151
|
Li JH, Ma J, Kang W, Wang CF, Bai F, Zhao K, Yao N, Liu Q, Dang BL, Wang BW, Wei QQ, Kang WZ, Sun YT. The histone deacetylase inhibitor chidamide induces intermittent viraemia in HIV-infected patients on suppressive antiretroviral therapy. HIV Med 2020; 21:747-757. [PMID: 33369029 DOI: 10.1111/hiv.13027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVES To evaluate the safety and efficacy of chidamide to reverse HIV-1 latency in vivo and to compare the effects of four clinically tested histone deacetylase (HDAC) inhibitors on non-histone proteins in vitro. METHODS Participants received chidamide orally at 10 mg twice weekly for 4 weeks while maintaining baseline antiretroviral therapy. The primary outcome was plasma viral rebound during chidamide dosing and the secondary outcomes were safety, pharmacokinetic and pharmacodynamic profiles, changes in cell-associated HIV-1 RNA and HIV-1 DNA, and immune parameters. Western blotting was used to compare the in vitro effects of the four HDAC inhibitors on HSP90, NF-κB and AP-1. RESULTS Seven aviraemic participants completed eight oral doses of chidamide, and only grade 1 adverse events were observed. Cyclic increases in histone acetylation were also detected. All participants showed robust and repeated plasma viral rebound (peak viraemia 147-3850 copies/mL), as well as increased cell-associated HIV-1 RNA, during chidamide treatment. Furthermore, we identified an enhanced HIV-1-specific cellular immune response and a modest 37.7% (95% CI: 12.7-62.8%, P = 0.028) reduction in cell-associated HIV-1 DNA. Compared with the other three HDAC inhibitors, chidamide had minimal cytotoxicity in vitro at clinically relevant concentrations and showed mechanistically superior effects on non-histone proteins, including HSP90, NF-κB and AP-1. CONCLUSIONS Chidamide safely and vigorously disrupts HIV-1 latency in vivo, which makes it a promising latency-reversing agent.
Collapse
Affiliation(s)
- J H Li
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - J Ma
- Department of Gastroenterology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - W Kang
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - C F Wang
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - F Bai
- Department of Infectious Diseases, 986 Hospital of Air Force affiliated to Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - K Zhao
- Department of Infectious Diseases, 986 Hospital of Air Force affiliated to Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - N Yao
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Q Liu
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - B L Dang
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - B W Wang
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Q Q Wei
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - W Z Kang
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Y T Sun
- Department of Infectious Diseases, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| |
Collapse
|
152
|
Prolonged administration of maraviroc reactivates latent HIV in vivo but it does not prevent antiretroviral-free viral rebound. Sci Rep 2020; 10:22286. [PMID: 33339855 PMCID: PMC7749169 DOI: 10.1038/s41598-020-79002-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/27/2020] [Indexed: 01/24/2023] Open
Abstract
Human immunodeficiency virus (HIV) remains incurable due to latent viral reservoirs established in non-activated CD4 T cells that cannot be eliminated via antiretroviral therapy. Current efforts to cure HIV are focused on identifying drugs that will induce viral gene expression in latently infected cells, commonly known as latency reversing agents (LRAs). Some drugs have been shown to reactivate latent HIV but do not cause a reduction in reservoir size. Therefore, finding new LRAs or new combinations or increasing the round of stimulations is needed to cure HIV. However, the effects of these drugs on viral rebound after prolonged treatment have not been evaluated. In a previous clinical trial, antiretroviral therapy intensification with maraviroc for 48 weeks caused an increase in residual viremia and episomal two LTR-DNA circles suggesting that maraviroc could reactivate latent HIV. We amended the initial clinical trial to explore additional virologic parameters in stored samples and to evaluate the time to viral rebound during analytical treatment interruption in three patients. Maraviroc induced an increase in cell-associated HIV RNA during the administration of the drug. However, there was a rapid rebound of viremia after antiretroviral therapy discontinuation. HIV-specific T cell response was slightly enhanced. These results show that maraviroc can reactivate latent HIV in vivo but further studies are required to efficiently reduce the reservoir size.
Collapse
|
153
|
Epigenetic Compound Screening Uncovers Small Molecules for Reactivation of Latent HIV-1. Antimicrob Agents Chemother 2020; 65:AAC.01815-20. [PMID: 33139279 DOI: 10.1128/aac.01815-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/22/2020] [Indexed: 11/20/2022] Open
Abstract
During infection with the human immunodeficiency virus type 1 (HIV-1), latent reservoirs are established that circumvent full eradication of the virus by antiretroviral therapy (ART) and are the source for viral rebound after cessation of therapy. As these reservoirs are phenotypically indistinguishable from infected cells, current strategies aim to reactivate these reservoirs, followed by pharmaceutical and immunological destruction of the cells. Here, we employed a simple and convenient cell-based reporter system, which enables sample handling under biosafety level (BSL)-1 conditions, to screen for compounds that were able to reactivate latent HIV-1. The assay showed a high dynamic signal range and reproducibility with an average Z-factor of 0.77, classifying the system as robust. The assay was used for high-throughput screening (HTS) of an epigenetic compound library in combination with titration and cell-toxicity studies and revealed several potential new latency-reversing agents (LRAs). Further validation in well-known latency model systems verified earlier studies and identified two novel compounds with very high reactivation efficiencies and low toxicity. Both drugs, namely, N-hydroxy-4-(2-[(2-hydroxyethyl)(phenyl)amino]-2-oxoethyl)benzamide (HPOB) and 2',3'-difluoro-[1,1'-biphenyl]-4-carboxylic acid, 2-butylhydrazide (SR-4370), showed comparable performances to other already known LRAs, did not activate CD4+ T cells, and did not cause changes in the composition of peripheral blood mononuclear cells (PBMCs), as shown by flow cytometry analyses. Both compounds may represent effective new treatment possibilities for reversal of latency in HIV-1-infected individuals.
Collapse
|
154
|
Moranguinho I, Valente ST. Block-And-Lock: New Horizons for a Cure for HIV-1. Viruses 2020; 12:v12121443. [PMID: 33334019 PMCID: PMC7765451 DOI: 10.3390/v12121443] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
HIV-1/AIDS remains a global public health problem. The world health organization (WHO) reported at the end of 2019 that 38 million people were living with HIV-1 worldwide, of which only 67% were accessing antiretroviral therapy (ART). Despite great success in the clinical management of HIV-1 infection, ART does not eliminate the virus from the host genome. Instead, HIV-1 remains latent as a viral reservoir in any tissue containing resting memory CD4+ T cells. The elimination of these residual proviruses that can reseed full-blown infection upon treatment interruption remains the major barrier towards curing HIV-1. Novel approaches have recently been developed to excise or disrupt the virus from the host cells (e.g., gene editing with the CRISPR-Cas system) to permanently shut off transcription of the virus (block-and-lock and RNA interference strategies), or to reactivate the virus from cell reservoirs so that it can be eliminated by the immune system or cytopathic effects (shock-and-kill strategy). Here, we will review each of these approaches, with the major focus placed on the block-and-lock strategy.
Collapse
|
155
|
Kleinman AJ, Xu C, Cottrell ML, Sivanandham R, Brocca-Cofano E, Dunsmore T, Kashuba A, Pandrea I, Apetrei C. Pharmacokinetics and Immunological Effects of Romidepsin in Rhesus Macaques. Front Immunol 2020; 11:579158. [PMID: 33362765 PMCID: PMC7759686 DOI: 10.3389/fimmu.2020.579158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
HIV/SIV persistence in latent reservoirs requires lifelong antiretroviral treatment and calls for effective cure strategies. Romidepsin (RMD), a histone deacetylase inhibitor, was reported to reactivate HIV/SIV from reservoirs in virus-suppressed individuals. We characterized in detail the pharmacokinetics and safety profile of RMD in three SIV-naïve rhesus macaques which received two rounds of treatment. In plasma, RMD mean terminal half-life was 15.3 h. In comparison, RMD mean terminal half-life was much longer in tissues: 110 h in the lymph nodes (LNs) and 28 h in gastrointestinal tract. RMD administration was accompanied by transient liver and systemic toxicity. Isoflurane anesthesia induced near-immediate transient lymphopenia, which was further exacerbated and extended with the extensive immune modifications by RMD. The effect of RMD on circulating immune cells was complex: (i) slight increase in lymphocyte death rates; (ii) transient, robust increase in neutrophils; (iii) massive downregulation of lymphocyte surface markers; (iv) important migration of CD3+ T cells to the gut and LNs; and (v) hindrance to CD8+ T cell functionality, yet without reaching significance. Our results show that, in contrast to transient plasma concentrations, RMD has a long-term presence in tissues, with multiple immunomodulatory effects and minimal to moderate kidney, liver, and lymphocyte toxicities. As such, we concluded that RMD can be used for “shock and kill” approaches, preferentially in combination with other latency reversal agents or cytotoxic T lymphocyte boosting strategies with consideration taken for adverse effects.
Collapse
Affiliation(s)
- Adam J Kleinman
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cuiling Xu
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mackenzie L Cottrell
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Ranjit Sivanandham
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Egidio Brocca-Cofano
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tammy Dunsmore
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Angela Kashuba
- University of North Carolina Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Infectious Diseases and Immunology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
156
|
HIV Antibody Fc N-Linked Glycosylation Is Associated with Viral Rebound. Cell Rep 2020; 33:108502. [PMID: 33326789 DOI: 10.1016/j.celrep.2020.108502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
Changes in antibody glycosylation are linked to inflammation across several diseases. Alterations in bulk antibody galactosylation can predict rheumatic flares, act as a sensor for immune activation, predict gastric cancer relapse, track with biological age, shift with vaccination, change with HIV reservoir size on therapy, and decrease in HIV and HCV infections. However, whether changes in antibody Fc biology also track with reservoir rebound time remains unclear. The identification of a biomarker that could forecast viral rebound time could significantly accelerate the downselection and iterative improvement of promising HIV viral eradication strategies. Using a comprehensive antibody Fc-profiling approach, the level of HIV-specific antibody Fc N-galactosylation is significantly associated with time to rebound after treatment discontinuation across three independent cohorts. Thus virus-specific antibody glycosylation may represent a promising, simply measured marker to track reservoir reactivation.
Collapse
|
157
|
Inderbitzin A, Kok YL, Jörimann L, Kelley A, Neumann K, Heinzer D, Cathomen T, Metzner KJ. HIV-1 promoter is gradually silenced when integrated into BACH2 in Jurkat T-cells. PeerJ 2020; 8:e10321. [PMID: 33282555 PMCID: PMC7694569 DOI: 10.7717/peerj.10321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/17/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The persistence of the latent HIV-1 reservoir is a major obstacle to curing HIV-1 infection. HIV-1 integrates into the cellular genome and some targeted genomic loci are frequently detected in clonally expanded latently HIV-1 infected cells, for instance, the gene BTB domain and CNC homology 2 (BACH2). METHODS We investigated HIV-1 promoter activity after integration into specific sites in BACH2 in Jurkat T-cells. The HIV-1-based vector LTatCL[M] contains two fluorophores: (1) Cerulean, which reports the activity of the HIV-1 promoter and (2) mCherry driven by a constitutive promotor and flanked by genetic insulators. This vector was inserted into introns 2 and 5 of BACH2 of Jurkat T-cells via CRISPR/Cas9 technology in the same and convergent transcriptional orientation of BACH2, and into the genomic safe harbour AAVS1. Single cell clones representing active (Cerulean+/mCherry+) and inactive (Cerulean-/mCherry+) HIV-1 promoters were characterised. RESULTS Upon targeted integration of the 5.3 kb vector LTatCL[M] into BACH2, the HIV-1 promoter was gradually silenced as reflected by the decrease in Cerulean expression over a period of 162 days. Silenced HIV-1 promoters could be reactivated by TNF-α and Romidepsin. This observation was independent of the targeted intron and the transcriptional orientation. BACH2 mRNA and protein expression was not impaired by mono-allelic integration of LTatCL[M]. CONCLUSION Successful targeted integration of the HIV-1-based vector LTatCL[M] allows longitudinal analyses of HIV-1 promoter activity.
Collapse
Affiliation(s)
- Anne Inderbitzin
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, University of Zurich, Zurich, Switzerland
| | - Yik Lim Kok
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Lisa Jörimann
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, University of Zurich, Zurich, Switzerland
| | - Audrey Kelley
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, University of Zurich, Zurich, Switzerland
| | - Kathrin Neumann
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Daniel Heinzer
- Institute for Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Neuroscience Center Zurich Graduate School, University of Zurich, Zurich, Switzerland
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Karin J. Metzner
- Department of Infectious Diseases and Hospital Epidemiology, Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
158
|
Vollbrecht T, Angerstein AO, Menke B, Kumar NM, de Oliveira MF, Richman DD, Guatelli JC. Inconsistent reversal of HIV-1 latency ex vivo by antigens of HIV-1, CMV, and other infectious agents. Retrovirology 2020; 17:36. [PMID: 33228686 PMCID: PMC7684880 DOI: 10.1186/s12977-020-00545-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A reservoir of replication-competent but latent virus is the main obstacle to a cure for HIV-1 infection. Much of this reservoir resides in memory CD4 T cells. We hypothesized that these cells can be reactivated with antigens from HIV-1 and other common pathogens to reverse latency. RESULTS We obtained mononuclear cells from the peripheral blood of antiretroviral-treated patients with suppressed viremia. We tested pools of peptides and proteins derived from HIV-1 and from other pathogens including CMV for their ability to reverse latency ex vivo by activation of memory responses. We assessed activation of the CD4 T cells by measuring the up-regulation of cell-surface CD69. We assessed HIV-1 expression using two assays: a real-time PCR assay for virion-associated viral RNA and a droplet digital PCR assay for cell-associated, multiply spliced viral mRNA. Reversal of latency occurred in a minority of cells from some participants, but no single antigen induced HIV-1 expression ex vivo consistently. When reversal of latency was induced by a specific peptide pool or protein, the extent was proportionally greater than that of T cell activation. CONCLUSIONS In this group of patients in whom antiretroviral therapy was started during chronic infection, the latent reservoir does not appear to consistently reside in CD4 T cells of a predominant antigen-specificity. Peptide-antigens reversed HIV-1 latency ex vivo with modest and variable activity. When latency was reversed by specific peptides or proteins, it was proportionally greater than the extent of T cell activation, suggesting partial enrichment of the latent reservoir in cells of specific antigen-reactivity.
Collapse
Affiliation(s)
- Thomas Vollbrecht
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- VA San Diego Healthcare System, San Diego, CA, USA.
| | - Aaron O Angerstein
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Bryson Menke
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Nikesh M Kumar
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Michelli Faria de Oliveira
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Douglas D Richman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - John C Guatelli
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
159
|
Replicate Aptima Assay for Quantifying Residual Plasma Viremia in Individuals on Antiretroviral Therapy. J Clin Microbiol 2020; 58:JCM.01400-20. [PMID: 32967900 PMCID: PMC7685884 DOI: 10.1128/jcm.01400-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022] Open
Abstract
Detection of residual plasma viremia in antiretroviral therapy (ART)-suppressed HIV-infected individuals is critical for characterizing the latent reservoir and evaluating the impact of cure interventions. Ultracentrifugation-based single-copy assays are sensitive but labor intensive. Fully automated replicate testing using a standard clinical viral load assay was evaluated as a high-throughput alternative for the quantification of low-level viremia. Four plasma samples from blood donors with acute HIV-1 infection and one viral culture supernatant were serially diluted into 25-ml samples to nominal viral loads ranging from 39 to <0. Detection of residual plasma viremia in antiretroviral therapy (ART)-suppressed HIV-infected individuals is critical for characterizing the latent reservoir and evaluating the impact of cure interventions. Ultracentrifugation-based single-copy assays are sensitive but labor intensive. Fully automated replicate testing using a standard clinical viral load assay was evaluated as a high-throughput alternative for the quantification of low-level viremia. Four plasma samples from blood donors with acute HIV-1 infection and one viral culture supernatant were serially diluted into 25-ml samples to nominal viral loads ranging from 39 to <0.5 copies (cp)/ml. Each dilution was tested with 45 replicates (reps) using 0.5 ml/rep with the Aptima HIV-1 Quant assay. The nominal and estimated viral loads based on the single-hit Poisson model were compared, and a hybrid Poisson digital model for calibrated viral load estimation was derived. Testing performed using 45 reps on longitudinal plasma samples from 50 ART-suppressed individuals in the Reservoir Assay Validation and Evaluation Network (RAVEN) study cohort (range of 1 to 19 years of continuous ART suppression) showed a median viral load of 0.54 cp/ml (interquartile range [IQR], 0.22 to 1.46 cp/ml) and a 14% (95% confidence interval [CI], 9% to 19%) decline in viral load for each additional year in duration suppressed. Within the RAVEN cohort, the expected false-negative rate for detection at lower rep numbers using 9 and 18 reps was 26% and 14%, respectively. Residual plasma viremia levels positively correlated with cell-associated HIV RNA and DNA. The performance characteristics of the replicate Aptima assay support its use for quantifying residual plasma viremia to study the latent HIV reservoir and cure interventions.
Collapse
|
160
|
TELWATTE S, KIM P, CHEN TH, MILUSH JM, SOMSOUK M, DEEKS SG, HUNT PW, WONG JK, YUKL SA. Mechanistic differences underlying HIV latency in the gut and blood contribute to differential responses to latency-reversing agents. AIDS 2020; 34:2013-2024. [PMID: 32910065 PMCID: PMC7990078 DOI: 10.1097/qad.0000000000002684] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE While latently HIV-infected cells have been described in the blood, it is unclear whether a similar inducible reservoir exists in the gut, where most HIV-infected cells reside. Tissue-specific environments may contribute to differences in the mechanisms that govern latent HIV infection and amenability to reactivation. We sought to determine whether HIV-infected cells from the blood and gut differ in their responses to T-cell activation and mechanistically distinct latency reversing agents (LRAs). DESIGN Cross sectional study using samples from HIV-infected individuals (n = 11). METHODS Matched peripheral blood mononuclear cells (PBMC) and dissociated total cells from rectum ± ileum were treated ex vivo for 24 h with anti-CD3/CD28 or LRAs in the presence of antiretrovirals. HIV DNA and 'read-through', initiated, 5' elongated, completed, and multiply-spliced HIV transcripts were quantified using droplet digital PCR. RESULTS T-cell activation increased levels of all HIV transcripts in PBMC and gut cells, and was the only treatment that increased multiply-spliced HIV RNA. Disulfiram increased initiated HIV transcripts in PBMC but not gut cells, while ingenol mebutate increased HIV transcription more in gut cells. Romidepsin increased HIV transcription in PBMC and gut cells, but the increase in transcription initiation was greater in PBMC. CONCLUSION The gut harbors HIV-infected cells in a latent-like state that can be reversed by T-cell activation involving CD3/CD28 signaling. Histone deacetylation and protein kinase B may contribute less to HIV transcriptional initiation in the gut, whereas protein kinase C may contribute more. New LRAs or combinations are needed to induce multiply-spliced HIV and should be tested on both blood and gut.
Collapse
Affiliation(s)
- Sushama TELWATTE
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA, 94121, USA
| | - Peggy KIM
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA, 94121, USA
| | - Tsui-Hua CHEN
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
| | - Jeffrey M. MILUSH
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
| | - Ma SOMSOUK
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
| | - Steven G. DEEKS
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
| | - Peter W. HUNT
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
| | - Joseph K. WONG
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA, 94121, USA
| | - Steven A. YUKL
- Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, 94110, USA
- Department of Medicine, San Francisco VA Medical Center, San Francisco, CA, 94121, USA
| |
Collapse
|
161
|
Sahay B, Mergia A. The Potential Contribution of Caveolin 1 to HIV Latent Infection. Pathogens 2020; 9:pathogens9110896. [PMID: 33121153 PMCID: PMC7692328 DOI: 10.3390/pathogens9110896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
Combinatorial antiretroviral therapy (cART) suppresses HIV replication to undetectable levels and has been effective in prolonging the lives of HIV infected individuals. However, cART is not capable of eradicating HIV from infected individuals mainly due to HIV’s persistence in small reservoirs of latently infected resting cells. Latent infection occurs when the HIV-1 provirus becomes transcriptionally inactive and several mechanisms that contribute to the silencing of HIV transcription have been described. Despite these advances, latent infection remains a major hurdle to cure HIV infected individuals. Therefore, there is a need for more understanding of novel mechanisms that are associated with latent infection to purge HIV from infected individuals thoroughly. Caveolin 1(Cav-1) is a multifaceted functional protein expressed in many cell types. The expression of Cav-1 in lymphocytes has been controversial. Recent evidence, however, convincingly established the expression of Cav-1 in lymphocytes. In lieu of this finding, the current review examines the potential role of Cav-1 in HIV latent infection and provides a perspective that helps uncover new insights to understand HIV latent infection.
Collapse
Affiliation(s)
| | - Ayalew Mergia
- Correspondence: ; Tel.: +352-294-4139; Fax: +352-392-9704
| |
Collapse
|
162
|
Latency-Reversing Agents Induce Differential Responses in Distinct Memory CD4 T Cell Subsets in Individuals on Antiretroviral Therapy. Cell Rep 2020; 29:2783-2795.e5. [PMID: 31775045 DOI: 10.1016/j.celrep.2019.10.101] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/11/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Latent proviruses persist in central (TCM), transitional (TTM), and effector (TEM) memory cells. We measured the levels of cellular factors involved in HIV gene expression in these subsets. The highest levels of acetylated H4, active nuclear factor κB (NF-κB), and active positive transcription elongation factor b (P-TEFb) were measured in TEM, TCM, and TTM cells, respectively. Vorinostat and romidepsin display opposite abilities to induce H4 acetylation across subsets. Protein kinase C (PKC) agonists are more efficient at inducing NF-κB phosphorylation in TCM cells but more potent at activating PTEF-b in the TEM subset. We selected the most efficient latency-reversing agents (LRAs) and measured their ability to reverse latency in each subset. While ingenol alone has modest activities in the three subsets, its combination with a histone deacetylase inhibitor (HDACi) dramatically increases latency reversal in TCM cells. Altogether, these results indicate that cellular HIV reservoirs are differentially responsive to common LRAs and suggest that combination of compounds will be required to achieve latency reversal in all subsets.
Collapse
|
163
|
French AJ, Natesampillai S, Krogman A, Correia C, Peterson KL, Alto A, Chandrasekar AP, Misra A, Li Y, Kaufmann SH, Badley AD, Cummins NW. Reactivating latent HIV with PKC agonists induces resistance to apoptosis and is associated with phosphorylation and activation of BCL2. PLoS Pathog 2020; 16:e1008906. [PMID: 33075109 PMCID: PMC7595626 DOI: 10.1371/journal.ppat.1008906] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/29/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Eradication of HIV-1 by the "kick and kill" strategy requires reactivation of latent virus to cause death of infected cells by either HIV-induced or immune-mediated apoptosis. To date this strategy has been unsuccessful, possibly due to insufficient cell death in reactivated cells to effectively reduce HIV-1 reservoir size. As a possible cause for this cell death resistance, we examined whether leading latency reversal agents (LRAs) affected apoptosis sensitivity of CD4 T cells. Multiple LRAs of different classes inhibited apoptosis in CD4 T cells. Protein kinase C (PKC) agonists bryostatin-1 and prostratin induced phosphorylation and enhanced neutralizing capability of the anti-apoptotic protein BCL2 in a PKC-dependent manner, leading to resistance to apoptosis induced by both intrinsic and extrinsic death stimuli. Furthermore, HIV-1 producing CD4 T cells expressed more BCL2 than uninfected cells, both in vivo and after ex vivo reactivation. Therefore, activation of BCL2 likely contributes to HIV-1 persistence after latency reversal with PKC agonists. The effects of LRAs on apoptosis sensitivity should be considered in designing HIV cure strategies predicated upon the "kick and kill" paradigm.
Collapse
Affiliation(s)
- Andrea J. French
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sekar Natesampillai
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ashton Krogman
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Cristina Correia
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kevin L. Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alecia Alto
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aswath P. Chandrasekar
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anisha Misra
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ying Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Scott H. Kaufmann
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Andrew D. Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Nathan W. Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
164
|
Warren JA, Zhou S, Xu Y, Moeser MJ, MacMillan DR, Council O, Kirchherr J, Sung JM, Roan NR, Adimora AA, Joseph S, Kuruc JD, Gay CL, Margolis DM, Archin N, Brumme ZL, Swanstrom R, Goonetilleke N. The HIV-1 latent reservoir is largely sensitive to circulating T cells. eLife 2020; 9:57246. [PMID: 33021198 PMCID: PMC7593086 DOI: 10.7554/elife.57246] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/24/2020] [Indexed: 01/01/2023] Open
Abstract
HIV-1-specific CD8+ T cells are an important component of HIV-1 curative strategies. Viral variants in the HIV-1 reservoir may limit the capacity of T cells to detect and clear virus-infected cells. We investigated the patterns of T cell escape variants in the replication-competent reservoir of 25 persons living with HIV-1 (PLWH) durably suppressed on antiretroviral therapy (ART). We identified all reactive T cell epitopes in the HIV-1 proteome for each participant and sequenced HIV-1 outgrowth viruses from resting CD4+ T cells. All non-synonymous mutations in reactive T cell epitopes were tested for their effect on the size of the T cell response, with a≥50% loss defined as an escape mutation. The majority (68%) of T cell epitopes harbored no detectable escape mutations. These findings suggest that circulating T cells in PLWH on ART could contribute to control of rebound and could be targeted for boosting in curative strategies.
Collapse
Affiliation(s)
- Joanna A Warren
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States
| | - Shuntai Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, United States.,UNC Center For AIDS Research, University of North Carolina, Chapel Hill, United States
| | - Yinyan Xu
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States
| | - Matthew J Moeser
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, United States.,UNC Center For AIDS Research, University of North Carolina, Chapel Hill, United States
| | | | - Olivia Council
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, United States
| | - Jennifer Kirchherr
- Department of Medicine, University of North Carolina, Chapel Hill, United States
| | - Julia M Sung
- Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - Nadia R Roan
- Department of Urology, University of California San Francisco, San Francisco, United States.,Gladstone Institute of Virology and Immunology, San Francisco, United States
| | - Adaora A Adimora
- Department of Medicine, University of North Carolina, Chapel Hill, United States
| | - Sarah Joseph
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - JoAnn D Kuruc
- Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - Cynthia L Gay
- Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - David M Margolis
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States.,UNC Center For AIDS Research, University of North Carolina, Chapel Hill, United States.,Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - Nancie Archin
- Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - Zabrina L Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada.,Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Ronald Swanstrom
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, United States.,UNC Center For AIDS Research, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| | - Nilu Goonetilleke
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, United States.,Department of Medicine, University of North Carolina, Chapel Hill, United States.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, United States
| |
Collapse
|
165
|
van Dorp CH, Conway JM, Barouch DH, Whitney JB, Perelson AS. Models of SIV rebound after treatment interruption that involve multiple reactivation events. PLoS Comput Biol 2020; 16:e1008241. [PMID: 33001979 PMCID: PMC7529301 DOI: 10.1371/journal.pcbi.1008241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
In order to assess the efficacy of novel HIV-1 treatments leading to a functional cure, the time to viral rebound is frequently used as a surrogate endpoint. The longer the time to viral rebound, the more efficacious the therapy. In support of such an approach, mathematical models serve as a connection between the size of the latent reservoir and the time to HIV-1 rebound after treatment interruption. The simplest of such models assumes that a single successful latent cell reactivation event leads to observable viremia after a period of exponential viral growth. Here we consider a generalization developed by Pinkevych et al. and Hill et al. of this simple model in which multiple reactivation events can occur, each contributing to the exponential growth of the viral load. We formalize and improve the previous derivation of the dynamics predicted by this model, and use the model to estimate relevant biological parameters from SIV rebound data. We confirm a previously described effect of very early antiretroviral therapy (ART) initiation on the rate of recrudescence and the viral load growth rate after treatment interruption. We find that every day ART initiation is delayed results in a 39% increase in the recrudescence rate (95% credible interval: [18%, 62%]), and a 11% decrease of the viral growth rate (95% credible interval: [4%, 20%]). We show that when viral rebound occurs early relative to the viral load doubling time, a model with multiple successful reactivation events fits the data better than a model with only a single successful reactivation event.
Collapse
Affiliation(s)
- Christiaan H. van Dorp
- Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jessica M. Conway
- Department of Mathematics and Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - James B. Whitney
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Alan S. Perelson
- Theoretical Biology and Biophysics (T-6), Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| |
Collapse
|
166
|
Campos Coelho AV, de Moura RR, Crovella S. Reanalysis of Gene Expression Profiles of CD4+ T Cells Treated with HIV-1 Latency Reversal Agents. Microorganisms 2020; 8:microorganisms8101505. [PMID: 33007800 PMCID: PMC7601709 DOI: 10.3390/microorganisms8101505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
The human immunodeficiency virus (HIV-1) causes a progressive depletion of CD4+ T cells, hampering immune function. Current experimental strategies to fight the virus focus on the reactivation of latent HIV-1 in the viral reservoir to make the virus detectable by the immune system, by searching for latency reversal agents (LRAs). We hypothesize that if common molecular pathways elicited by the presence of LRAs are known, perhaps new, more efficient, “shock-and-kill” strategies can be found. Thus, the objective of the present study is to re-evaluate RNA-Seq assays to find differentially expressed genes (DEGs) during latency reversal via transcriptome analysis. We selected six studies (45 samples altogether: 16 negative controls and 29 LRA-treated CD4+ T cells) and 11 LRA strategies through a systematic search in Gene Expression Omnibus (GEO) and PubMed databases. The raw reads were trimmed, counted, and normalized. Next, we detected consistent DEGs in these independent experiments. AZD5582, romidepsin, and suberanilohydroxamic acid (SAHA) were the LRAs that modulated most genes. We detected 948 DEGs shared by those three LRAs. Gene ontology analysis and cross-referencing with other sources of the literature showed enrichment of cell activation, differentiation and signaling, especially mitogen-activated protein kinase (MAPK) and Rho-GTPases pathways.
Collapse
Affiliation(s)
- Antonio Victor Campos Coelho
- Federal University of Pernambuco, Avenida da Engenharia, Cidade Universitária, Recife 50670-901, Brazil
- Correspondence: ; Tel.: +55-81-2126-8522
| | - Ronald Rodrigues de Moura
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (R.R.d.M.); (S.C.)
| | - Sergio Crovella
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (R.R.d.M.); (S.C.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
167
|
Ventura JD. Human Immunodeficiency Virus 1 (HIV-1): Viral Latency, the Reservoir, and the Cure. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:549-560. [PMID: 33005119 PMCID: PMC7513431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An estimated 37 million people globally suffer from Human Immunodeficiency Virus-1 (HIV-1) infection with 1.7 million newly acquired infections occurring on average each year. Although crucial advances in combined antiretroviral therapy (ART) over the last two decades have transformed an HIV-1 diagnosis into a tolerable and controlled condition, enabling over 20 million people living with HIV-1 to enjoy healthy and productive lives, no cure or vaccine yet exists. Developing a successful cure strategy will require a firm understanding of how viral latency is established and how a persistent and long-lived latent is generated. The latent reservoir remains the primary obstacle for cure development and most putative cure strategies proposed fundamentally address its eradication or permanent suppression.
Collapse
Affiliation(s)
- John D. Ventura
- To whom all correspondence should be addressed:
Dr. John D. Ventura, . ORCID iD:
https://orcid.org/0000-0002-4373-3242.
| |
Collapse
|
168
|
Ward AR, Mota TM, Jones RB. Immunological approaches to HIV cure. Semin Immunol 2020; 51:101412. [PMID: 32981836 DOI: 10.1016/j.smim.2020.101412] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Combination antiretroviral therapy (ART) to treat human immunodeficiency virus (HIV) infection has proven remarkably successful - for those who can access and afford it - yet HIV infection persists indefinitely in a reservoir of cells, despite effective ART and despite host antiviral immune responses. An HIV cure is therefore the next aspirational goal and challenge, though approaches differ in their objectives - with 'functional cures' aiming for durable viral control in the absence of ART, and 'sterilizing cures' aiming for the more difficult to realize objective of complete viral eradication. Mechanisms of HIV persistence, including viral latency, anatomical sequestration, suboptimal immune functioning, reservoir replenishment, target cell-intrinsic immune resistance, and, potentially, target cell distraction of immune effectors, likely need to be overcome in order to achieve a cure. A small fraction of people living with HIV (PLWH) naturally control infection via immune-mediated mechanisms, however, providing both sound rationale and optimism that an immunological approach to cure is possible. Herein we review up to date knowledge and emerging evidence on: the mechanisms contributing to HIV persistence, as well as potential strategies to overcome these barriers; promising immunological approaches to achieve viral control and elimination of reservoir-harboring cells, including harnessing adaptive immune responses to HIV and engineered therapies, as well as enhancers of their functions and of complementary innate immune functioning; and combination strategies that are most likely to succeed. Ultimately, a cure must be safe, effective, durable, and, eventually, scalable in order to be widely acceptable and available.
Collapse
Affiliation(s)
- Adam R Ward
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA; PhD Program in Epidemiology, The George Washington University, Washington, DC, USA
| | - Talia M Mota
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - R Brad Jones
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
169
|
López-Huertas MR, Jiménez-Tormo L, Madrid-Elena N, Gutiérrez C, Vivancos MJ, Luna L, Moreno S. Maraviroc reactivates HIV with potency similar to that of other latency reversing drugs without inducing toxicity in CD8 T cells. Biochem Pharmacol 2020; 182:114231. [PMID: 32979351 DOI: 10.1016/j.bcp.2020.114231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/21/2020] [Indexed: 01/12/2023]
Abstract
Human immunodeficiency virus (HIV) remains incurable due to latent reservoirs established in non-activated CD4 T cells. Current efforts to achieve a functional cure rely on immunomodulatory strategies focused on enhancing the functions of cytotoxic cells. Implementation of these actions requires a coordinated activation of the viral transcription in latently infected cells so that the reservoir became visible and accessible to cytotoxic cells. As no latency reversing agent (LRA) has been shown to be completely effective, new combinations are of increasing importance. Recent data have shown that maraviroc is a new LRA. In this work, we have explored how the combination of maraviroc with other LRAs influences on HIV reactivation using in vitro latency models as well as on the cell viability of CD8 T cells from ART-treated patients. Maraviroc reactivated HIV with a potency similar to other LRAs. Triple combinations resulted toxic and were rejected. No dual combination was synergistic. The combination with panobinostat or disulfiram maintained the effect of both drugs without inducing cell proliferation or toxicity. Maraviroc does not alter the viability of CD8 T cells isolated from patients under antiretroviral treatment. This finding enhances the properties of maraviroc as a LRA.
Collapse
Affiliation(s)
- María Rosa López-Huertas
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain.
| | - Laura Jiménez-Tormo
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Nadia Madrid-Elena
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Carolina Gutiérrez
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - María Jesús Vivancos
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Laura Luna
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Santiago Moreno
- Servicio de Enfermedades Infecciosas, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain; Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá de Henares, 28871 Alcalá de Henares, Spain
| |
Collapse
|
170
|
Combination of CD8β Depletion and Interleukin-15 Superagonist N-803 Induces Virus Reactivation in Simian-Human Immunodeficiency Virus-Infected, Long-Term ART-Treated Rhesus Macaques. J Virol 2020; 94:JVI.00755-20. [PMID: 32669328 PMCID: PMC7495383 DOI: 10.1128/jvi.00755-20] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The “shock and kill” HIV cure strategy attempts to reverse and eliminate the latent viral infection that prevents eradication of the virus. Latency-reversing agents tested in clinical trials to date have failed to affect the HIV viral reservoir. IL-15 superagonist N-803, currently involved in a clinical trial for HIV cure, was recently shown by our laboratory to induce robust and persistent induction of plasma viremia during ART in three in vivo animal models of HIV infection. These results suggest a substantial role for CD8+ lymphocytes in suppressing the latency reversal effect of N-803 by promoting the maintenance of viral latency. In this study, we tested whether the use of a CD8β-targeting antibody, which would specifically deplete CD8+ T cells, would yield similar levels of virus reactivation. We observed the induction of plasma viremia, which correlated with the efficacy of the CD8 depletion strategy. The “shock and kill” strategy predicates that virus reactivation in latently infected cells is required to eliminate the human immunodeficiency virus (HIV) reservoir. In a recent study, we showed robust and persistent induction of plasma viremia in antiretroviral therapy (ART)-treated simian immunodeficiency virus-infected rhesus macaques (RMs) undergoing CD8α depletion and treated with the interleukin-15 (IL-15) superagonist N-803 (J. B. McBrien et al., Nature 578:154–159, 2020, https://doi.org/10.1038/s41586-020-1946-0). Of note, in that study we used an antibody targeting CD8α, thereby depleting NK cells, NKT cells, and γδ T cells, in addition to CD8+ T cells. In the current proof-of-concept study, we tested whether virus reactivation can be induced by administration of N-803 to simian-human chimeric immunodeficiency virus-infected, ART-treated RMs that are selectively depleted of CD8+ T cells via the CD8β-targeting antibody CD8b255R1. CD8β depletion was performed in five SHIVSF162P3-infected RMs treated with ART for 12 months and with plasma viremia consistently below 3 copies/ml. All animals received four weekly doses of N-803 starting at the time of CD8b255R1 administration. The induction of detectable plasma viremia was observed in three out of five RMs, with the level of virus reactivation seemingly correlated with the frequency of CD8+ T cells following CD8β depletion as well as the level of virus reactivation observed when the same animals underwent CD8α depletion and N-803 administration after 24 weeks of ART. These data indicate that CD8β depletion and N-803 administration can induce virus reactivation in SHIVSF162P3-infected RMs despite suboptimal depletion of CD8+ T cells and profound ART-induced suppression of virus replication, confirming a critical role for these cells in suppressing virus production and/or reactivation in vivo under ART. IMPORTANCE The “shock and kill” HIV cure strategy attempts to reverse and eliminate the latent viral infection that prevents eradication of the virus. Latency-reversing agents tested in clinical trials to date have failed to affect the HIV viral reservoir. IL-15 superagonist N-803, currently involved in a clinical trial for HIV cure, was recently shown by our laboratory to induce robust and persistent induction of plasma viremia during ART in three in vivo animal models of HIV infection. These results suggest a substantial role for CD8+ lymphocytes in suppressing the latency reversal effect of N-803 by promoting the maintenance of viral latency. In this study, we tested whether the use of a CD8β-targeting antibody, which would specifically deplete CD8+ T cells, would yield similar levels of virus reactivation. We observed the induction of plasma viremia, which correlated with the efficacy of the CD8 depletion strategy.
Collapse
|
171
|
Yang X, Wang Y, Lu P, Shen Y, Zhao X, Zhu Y, Jiang Z, Yang H, Pan H, Zhao L, Zhong Y, Wang J, Liang Z, Shen X, Lu D, Jiang S, Xu J, Wu H, Lu H, Jiang G, Zhu H. PEBP1 suppresses HIV transcription and induces latency by inactivating MAPK/NF-κB signaling. EMBO Rep 2020; 21:e49305. [PMID: 32924251 PMCID: PMC7645261 DOI: 10.15252/embr.201949305] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 05/28/2020] [Accepted: 08/12/2020] [Indexed: 11/09/2022] Open
Abstract
The latent HIV‐1 reservoir is a major barrier to viral eradication. However, our understanding of how HIV‐1 establishes latency is incomplete. Here, by performing a genome‐wide CRISPR‐Cas9 knockout library screen, we identify phosphatidylethanolamine‐binding protein 1 (PEBP1), also known as Raf kinase inhibitor protein (RKIP), as a novel gene inducing HIV latency. Depletion of PEBP1 leads to the reactivation of HIV‐1 in multiple models of latency. Mechanistically, PEBP1 de‐phosphorylates Raf1/ERK/IκB and IKK/IκB signaling pathways to sequestrate NF‐κB in the cytoplasm, which transcriptionally inactivates HIV‐1 to induce latency. Importantly, the induction of PEBP1 expression by the green tea compound epigallocatechin‐3‐gallate (EGCG) prevents latency reversal by inhibiting nuclear translocation of NF‐κB, thereby suppressing HIV‐1 transcription in primary CD4+ T cells isolated from patients receiving antiretroviral therapy (ART). These results suggest a critical role for PEBP1 in the regulation of upstream NF‐κB signaling pathways governing HIV transcription. Targeting of this pathway could be an option to control HIV reservoirs in patients in the future.
Collapse
Affiliation(s)
- Xinyi Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanan Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Panpan Lu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yinzhong Shen
- Department of Infectious Disease, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaying Zhao
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuqi Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhengtao Jiang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - He Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Hanyu Pan
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Lin Zhao
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yangcheng Zhong
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhiming Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoting Shen
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Shibo Jiang
- Department of Infectious Disease, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianqing Xu
- Department of Infectious Disease, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hao Wu
- Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Hongzhou Lu
- Department of Infectious Disease, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Guochun Jiang
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases & Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Huanzhang Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
172
|
Kroon ED, Ananworanich J, Pagliuzza A, Rhodes A, Phanuphak N, Trautmann L, Mitchell JL, Chintanaphol M, Intasan J, Pinyakorn S, Benjapornpong K, Chang JJ, Colby DJ, Chomchey N, Fletcher JL, Eubanks K, Yang H, Kapson J, Dantanarayana A, Tennakoon S, Gorelick RJ, Maldarelli F, Robb ML, Kim JH, Spudich S, Chomont N, Phanuphak P, Lewin SR, de Souza MS, for the SEARCH 019 and RV254 Study Teams. A randomized trial of vorinostat with treatment interruption after initiating antiretroviral therapy during acute HIV-1 infection. J Virus Erad 2020; 6:100004. [PMID: 33251022 PMCID: PMC7646672 DOI: 10.1016/j.jve.2020.100004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE AND DESIGN A randomized, open-label pilot study in individuals treated with antiretroviral therapy (ART) since acute HIV infection (AHI) with a regimen including a histone deacetylase inhibitor to induce HIV from latency and control HIV replication during subsequent treatment interruption (TI). METHODS Fifteen participants who initiated ART at AHI were randomized to vorinostat/hydroxychloroquine/maraviroc (VHM) plus ART (n = 10) or ART alone (n = 5). The VHM arm received three 14-day vorinostat cycles within 10 weeks before TI. ART was resumed for plasma viral load (VL) > 1,000 HIV RNA copies/mL. Primary outcome was proportion of participants on VHM + ART versus ART only with VL < 50 copies/mL for 24 weeks after TI. RESULTS Fifteen participants on ART (median: 178 weeks: range 79-295) enrolled. Two on VHM + ART experienced serious adverse events. Fourteen participants underwent TI; all experienced VL rebound with no difference in time between arms: VHM + ART (n = 9) median: 4 weeks and ART only (n = 5) median: 5 weeks. VHM induced a 2.2-fold increase in VL (p = 0.008) by single-copy HIV RNA assay after the first cycle. Neopterin levels increased significantly following the first two cycles. After VHM treatment, the frequencies of peripheral blood mononuclear cells harboring total HIV DNA and cell-associated RNA were unchanged. All participants achieved VL suppression following ART re-initiation. CONCLUSIONS Administration of VHM increased HIV VL in plasma, but this was not sustained. VHM did not impact time to viral rebound following TI and had no impact on the size of the HIV reservoir, suggesting that HIV reservoir elimination will require alternative treatment strategies.
Collapse
Affiliation(s)
| | - Jintanat Ananworanich
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
- Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Amélie Pagliuzza
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
| | - Ajantha Rhodes
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
| | | | - Lydie Trautmann
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
| | - Julie L. Mitchell
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
| | - Michelle Chintanaphol
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- Department of Neurology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Jintana Intasan
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Suteeraporn Pinyakorn
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
| | | | - J. Judy Chang
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
| | - Donn J. Colby
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Nitiya Chomchey
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | | | | | - Hua Yang
- Cooper Human Systems, Nashua, NH, USA
| | | | - Ashanti Dantanarayana
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
| | - Surekha Tennakoon
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
| | - Robert J. Gorelick
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Frank Maldarelli
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Merlin L. Robb
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
| | - Jerome H. Kim
- International Vaccine Initiative, Seoul, Republic of Korea
| | - Serena Spudich
- Department of Neurology, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Nicolas Chomont
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
| | | | - Sharon R. Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Australia
| | | | - for the SEARCH 019 and RV254 Study Teams
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- United States Military HIV Research Program, Bethesda, MD, USA
- Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
- Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Canada
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Center, Melbourne, Australia
- Department of Neurology, Yale University School of Medicine, Yale University, New Haven, CT, USA
- Cooper Human Systems, Nashua, NH, USA
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- International Vaccine Initiative, Seoul, Republic of Korea
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Australia
| |
Collapse
|
173
|
Piekna-Przybylska D, Bambara RA, Maggirwar SB, Dewhurst S. G-quadruplex ligands targeting telomeres do not inhibit HIV promoter activity and cooperate with latency reversing agents in killing latently infected cells. Cell Cycle 2020; 19:2298-2313. [PMID: 32807015 DOI: 10.1080/15384101.2020.1796268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Altered telomere maintenance mechanism (TMM) is linked to increased DNA damage at telomeres and telomere uncapping. We previously showed that HIV-1 latent cells have altered TMM and are susceptible to ligands that target G-quadruplexes (G4) at telomeres. Susceptibility of latent cells to telomere targeting could potentially be used to support approaches to eradicate HIV reservoirs. However, G4 ligands also target G-quadruplexes in promoters blocking gene transcription. Since HIV promoter sequence can form G-quadruplexes, we investigated whether G4 ligands interfere with HIV-1 promoter activity and virus reactivation from latency, and whether telomere targeting could be combined with latency reversing agents (LRAs) to promote elimination of HIV reservoirs. Our results indicate that Sp1 binding region in HIV-1 promoter can adopt G4 structures in duplex DNA, and that in vitro binding of Sp1 to G-quadruplex is blocked by G4 ligand, suggesting that agents targeting telomeres interfere with virus reactivation. However, our studies show that G4 agents do not affect HIV-1 promoter activity in cell culture, and do not interfere with latency reversal. Importantly, primary memory CD4 + T cells infected with latent HIV-1 are more susceptible to combined treatment with LRAs and G4 ligands, indicating that drugs targeting TMM may enhance killing of HIV reservoirs. Using a cell-based DNA repair assay, we also found that HIV-1 infected cells have reduced efficiency of DNA mismatch repair (MMR), and base excision repair (BER), suggesting that altered TMM in latently infected cells could be associated with accumulation of DNA damage at telomeres and changes in telomeric caps.
Collapse
Affiliation(s)
- Dorota Piekna-Przybylska
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| | - Robert A Bambara
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| | - Sanjay B Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University , Washington, DC, USA
| | - Stephen Dewhurst
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| |
Collapse
|
174
|
Mu W, Carrillo MA, Kitchen SG. Engineering CAR T Cells to Target the HIV Reservoir. Front Cell Infect Microbiol 2020; 10:410. [PMID: 32903563 PMCID: PMC7438537 DOI: 10.3389/fcimb.2020.00410] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022] Open
Abstract
The HIV reservoir remains to be a difficult barrier to overcome in order to achieve a therapeutic cure for HIV. Several strategies have been developed to purge the reservoir, including the “kick and kill” approach, which is based on the notion that reactivating the latent reservoir will allow subsequent elimination by the host anti-HIV immune cells. However, clinical trials testing certain classes of latency reactivating agents (LRAs) have so far revealed the minimal impact on reducing the viral reservoir. A robust immune response to reactivated HIV expressing cells is critical for this strategy to work. A current focus to enhance anti-HIV immunity is through the use of chimeric antigen receptors (CARs). Currently, HIV-specific CARs are being applied to peripheral T cells, NK cells, and stem cells to boost recognition and killing of HIV infected cells. In this review, we summarize current developments in engineering HIV directed CAR-expressing cells to facilitate HIV elimination. We also summarize current LRAs that enhance the “kick” strategy and how new generation and combinations of LRAs with HIV specific CAR T cell therapies could provide an optimal strategy to target the viral reservoir and achieve HIV clearance from the body.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayra A Carrillo
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Scott G Kitchen
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
175
|
Mobasheri T, Rayzan E, Shabani M, Hosseini M, Mahmoodi Chalbatani G, Rezaei N. Neuroblastoma-targeted nanoparticles and novel nanotechnology-based treatment methods. J Cell Physiol 2020; 236:1751-1775. [PMID: 32735058 DOI: 10.1002/jcp.29979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022]
Abstract
Neuroblastoma is a complicated pediatric tumor, originating from the neural crest, which is the most prevalent in adrenal glands, but may rarely be seen in some other tissues as well. Studies are focused on developing new strategies through novel chemo- and immuno-therapeutic drug targets. Different types of oncogenes such as MYCN, tumor suppressor genes such as p53, and some structural genes such as vascular endothelial growth factor are considered as targets for neuroblastoma therapy. The individual expression patterns in NB cells make them appropriate for this purpose. The combined effect of nano-drug delivery systems and specific drug targets will result in lower systemic side effects, prolonged therapeutic effects, and improvements in the pharmacokinetic properties of the drugs. Some of these novel drug delivery systems with a focus on liposomes as carriers are also discussed. In this review, genes and protein products that are beneficial as drug targets in the treatment of neuroblastoma have been discussed.
Collapse
Affiliation(s)
- Taranom Mobasheri
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsima Shabani
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Baltimore, Maryland
| | - Mina Hosseini
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nima Rezaei
- Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
176
|
Namdari H, Rezaei F, Teymoori-Rad M, Mortezagholi S, Sadeghi A, Akbari A. CAR T cells: Living HIV drugs. Rev Med Virol 2020; 30:1-14. [PMID: 32713110 DOI: 10.1002/rmv.2139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1), the virus that causes AIDS (acquired immunodeficiency syndrome), is a major global public health issue. Although the advent of combined antiretroviral therapy (ART) has made significant progress in inhibiting HIV replication in patients, HIV-infected cells remain the principal cellular reservoir of HIV, this allows HIV to rebound immediately upon stopping ART, which is considered the major obstacle to curing HIV infection. Chimeric antigen receptor (CAR) cell therapy has provided new opportunities for HIV treatment. Engineering T cells or hematopoietic stem cells (HSCs) to generate CAR T cells is a rapidly growing approach to develop an efficient immune cell to fight HIV. Herein, we review preclinical and clinical data available for the development of CAR T cells. Further, the advantages and disadvantages of clinical application of anti-HIV CAR T cells will be discussed.
Collapse
Affiliation(s)
- Haideh Namdari
- Iranian Tissue Bank Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Mortezagholi
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Sadeghi
- Iranian Tissue Bank Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
177
|
Zhuang X, Pedroza-Pacheco I, Nawroth I, Kliszczak AE, Magri A, Paes W, Rubio CO, Yang H, Ashcroft M, Mole D, Balfe P, Borrow P, McKeating JA. Hypoxic microenvironment shapes HIV-1 replication and latency. Commun Biol 2020; 3:376. [PMID: 32665623 PMCID: PMC7360605 DOI: 10.1038/s42003-020-1103-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Viral replication is defined by the cellular microenvironment and one key factor is local oxygen tension, where hypoxia inducible factors (HIFs) regulate the cellular response to oxygen. Human immunodeficiency virus (HIV) infected cells within secondary lymphoid tissues exist in a low-oxygen or hypoxic environment in vivo. However, the majority of studies on HIV replication and latency are performed under laboratory conditions where HIFs are inactive. We show a role for HIF-2α in restricting HIV transcription via direct binding to the viral promoter. Hypoxia reduced tumor necrosis factor or histone deacetylase inhibitor, Romidepsin, mediated reactivation of HIV and inhibiting HIF signaling-pathways reversed this phenotype. Our data support a model where the low-oxygen environment of the lymph node may suppress HIV replication and promote latency. We identify a mechanism that may contribute to the limited efficacy of latency reversing agents in reactivating HIV and suggest new strategies to control latent HIV-1.
Collapse
Affiliation(s)
- Xiaodong Zhuang
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | | | - Isabel Nawroth
- Institute of Immunity and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anna E Kliszczak
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Andrea Magri
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Wayne Paes
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | | | - Hongbing Yang
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK
| | - David Mole
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Peter Balfe
- Institute of Immunity and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Persephone Borrow
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Jane A McKeating
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
178
|
Liu J, Kelly J, Yu W, Clausen D, Yu Y, Kim H, Duffy JL, Chung CC, Myers RW, Carroll S, Klein DJ, Fells J, Holloway MK, Wu J, Wu G, Howell BJ, Barnard RJO, Kozlowski JA. Selective Class I HDAC Inhibitors Based on Aryl Ketone Zinc Binding Induce HIV-1 Protein for Clearance. ACS Med Chem Lett 2020; 11:1476-1483. [PMID: 32676157 PMCID: PMC7357218 DOI: 10.1021/acsmedchemlett.0c00302] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 12/27/2022] Open
Abstract
HIV persistence in latently infected, resting CD4+ T cells is broadly considered a barrier to eradicate HIV. Activation of the provirus using latency-reversing agents (LRAs) followed by immune-mediated clearance to purge reservoirs has been touted as a promising therapeutic approach. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) control the acetylation level of lysine residues in histones to regulate the gene transcription. Several clinical HDAC inhibitors had been examined as LRAs, which induced HIV activation in vitro and in vivo. Here we report the discovery of a series of selective and potent class I HDAC inhibitors based on aryl ketones as a zinc binding group, which reversed HIV latency using a Jurkat model of HIV latency in 2C4 cells. The SAR led to the discovery of a highly selective class I HDAC inhibitor 10 with excellent potency. HDACi 10 induces the HIV gag P24 protein in patient latent CD4+ T cells.
Collapse
Affiliation(s)
- Jian Liu
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Joseph Kelly
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Wensheng Yu
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Dane Clausen
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Younong Yu
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hyunjin Kim
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Joseph L. Duffy
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Christine C. Chung
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert W. Myers
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Steve Carroll
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Daniel J. Klein
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - James Fells
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - M. Katharine Holloway
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jin Wu
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Guoxin Wu
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Bonnie J. Howell
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Richard J. O. Barnard
- Merck
& Co., Inc., 770
Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Joseph A. Kozlowski
- Merck
& Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
179
|
Yu W, Liu J, Yu Y, Zhang V, Clausen D, Kelly J, Wolkenberg S, Beshore D, Duffy JL, Chung CC, Myers RW, Klein DJ, Fells J, Holloway K, Wu J, Wu G, Howell BJ, Barnard RJ, Kozlowski J. Discovery of ethyl ketone-based HDACs 1, 2, and 3 selective inhibitors for HIV latency reactivation. Bioorg Med Chem Lett 2020; 30:127197. [DOI: 10.1016/j.bmcl.2020.127197] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 11/17/2022]
|
180
|
Abstract
Although antiretroviral therapies (ARTs) potently inhibit HIV replication, they do not eradicate the virus. HIV persists in cellular and anatomical reservoirs that show minimal decay during ART. A large number of studies conducted during the past 20 years have shown that HIV persists in a small pool of cells harboring integrated and replication-competent viral genomes. The majority of these cells do not produce viral particles and constitute what is referred to as the latent reservoir of HIV infection. Therefore, although HIV is not considered as a typical latent virus, it can establish a state of nonproductive infection under rare circumstances, particularly in memory CD4+ T cells, which represent the main barrier to HIV eradication. While it was originally thought that the pool of latently infected cells was largely composed of cells harboring transcriptionally silent genomes, recent evidence indicates that several blocks contribute to the nonproductive state of these cells. Here, we describe the virological and immunological factors that play a role in the establishment and persistence of the pool of latently infected cells and review the current approaches aimed at eliminating the latent HIV reservoir.
Collapse
Affiliation(s)
| | - Pierre Gantner
- Department of Microbiology, Infectiology and Immunology and
| | - Rémi Fromentin
- Centre de Recherche du Centre Hospitalier, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology and
- Centre de Recherche du Centre Hospitalier, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
181
|
Lima NS, Takata H, Huang SH, Haregot A, Mitchell J, Blackmore S, Garland A, Sy A, Cartwright P, Routy JP, Michael NL, Appay V, Jones RB, Trautmann L. CTL Clonotypes with Higher TCR Affinity Have Better Ability to Reduce the HIV Latent Reservoir. THE JOURNAL OF IMMUNOLOGY 2020; 205:699-707. [PMID: 32591402 DOI: 10.4049/jimmunol.1900811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 05/23/2020] [Indexed: 01/09/2023]
Abstract
The success of the shock and kill strategy for the HIV cure depends both on the reactivation of the latent reservoir and on the ability of the immune system to eliminate infected cells. As latency reversal alone has not shown any impact in the size of the latent reservoir, ensuring that effector CTLs are able to recognize and kill HIV-infected cells could contribute to reservoir reduction. In this study, we investigated which functional aspects of human CTLs are associated with a better capacity to kill HIV-infected CD4+ T cells. We isolated Gag- and Nef-specific CTL clones with different TCR sequences from the PBMC of donors in acute and chronic infection. High-affinity clonotypes that showed IFN-γ production preserved even when the CD8 coreceptor was blocked, and clones with high Ag sensitivity exhibited higher efficiency at reducing the latent reservoir. Although intrinsic cytotoxic capacity did not differ according to TCR affinity, clonotypes with high TCR affinity showed a better ability to kill HIV-infected CD4+ T cells obtained from in vivo-infected PBMC and subjected to viral reactivation. Strategies aiming to specifically boost and maintain long-living memory CTLs with high TCR affinity in vivo prior to latency-reversing treatment might improve the efficacy of the shock and kill approach to reduce the latent reservoir.
Collapse
Affiliation(s)
- Noemia S Lima
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814
| | - Hiroshi Takata
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| | - Szu-Han Huang
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021.,Department of Microbiology, Immunology, and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Alexander Haregot
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Julie Mitchell
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| | - Stephen Blackmore
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Ayanna Garland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Aaron Sy
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | | | - Jean-Pierre Routy
- Division of Hematology and Chronic Viral Illness Service, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Nelson L Michael
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910
| | - Victor Appay
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Université, INSERM, Paris 75005, France; and.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto 860-8555, Japan
| | - R Brad Jones
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10021.,Department of Microbiology, Immunology, and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Lydie Trautmann
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910; .,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817.,Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006
| |
Collapse
|
182
|
Pache L, Marsden MD, Teriete P, Portillo AJ, Heimann D, Kim JT, Soliman MS, Dimapasoc M, Carmona C, Celeridad M, Spivak AM, Planelles V, Cosford ND, Zack JA, Chanda SK. Pharmacological Activation of Non-canonical NF-κB Signaling Activates Latent HIV-1 Reservoirs In Vivo. Cell Rep Med 2020; 1:100037. [PMID: 33205060 PMCID: PMC7659604 DOI: 10.1016/j.xcrm.2020.100037] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/01/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023]
Abstract
"Shock and kill" strategies focus on purging the latent HIV-1 reservoir by treating infected individuals with therapeutics that activate the latent virus and subsequently eliminating infected cells. We have previously reported that induction of non-canonical nuclear factor κB (NF-κB) signaling through a class of small-molecule antagonists known as Smac mimetics can reverse HIV-1 latency. Here, we describe the development of Ciapavir (SBI-0953294), a molecule specifically optimized for HIV-1 latency reversal that was found to be more efficacious as a latency-reversing agent than other Smac mimetics under clinical development for cancer. Critically, this molecule induced activation of HIV-1 reservoirs in vivo in a bone marrow, liver, thymus (BLT) humanized mouse model without mediating systemic T cell activation. This study provides proof of concept for the in vivo efficacy and safety of Ciapavir and indicates that Smac mimetics can constitute a critical component of a safe and efficacious treatment strategy to eliminate the latent HIV-1 reservoir.
Collapse
Affiliation(s)
- Lars Pache
- Infectious and Inflammatory Disease Center, Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Matthew D. Marsden
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peter Teriete
- Cell Metabolism and Signaling Networks Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alex J. Portillo
- Infectious and Inflammatory Disease Center, Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dominik Heimann
- Cell Metabolism and Signaling Networks Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jocelyn T. Kim
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mohamed S.A. Soliman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melanie Dimapasoc
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Camille Carmona
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maria Celeridad
- Cell Metabolism and Signaling Networks Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Adam M. Spivak
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Vicente Planelles
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Nicholas D.P. Cosford
- Cell Metabolism and Signaling Networks Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jerome A. Zack
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sumit K. Chanda
- Infectious and Inflammatory Disease Center, Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
183
|
Cellular Gene Modulation of HIV-Infected CD4 T Cells in Response to Serial Treatment with the Histone Deacetylase Inhibitor Vorinostat. J Virol 2020; 94:JVI.00351-20. [PMID: 32295913 PMCID: PMC7307144 DOI: 10.1128/jvi.00351-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 12/23/2022] Open
Abstract
Histone deacetylase inhibitors are widely studied HIV latency-reversing agents (LRAs). VOR, an HDACi, induces histone acetylation and chromatin remodeling and modulates host and HIV gene expression. However, the relationship between these events is poorly defined, and clinical studies suggest diminished HIV reactivation in resting CD4 T cells with daily exposure to VOR. Our study provides evidence that VOR induces a consistent level of host cell gene transcription following intermittent exposure. In addition, in response to VOR exposure a gene signature that was conserved across single and serial exposures both in vitro and in vivo was identified, indicating that VOR can consistently and reproducibly modulate transcriptional host responses. However, as the HIV response to HDACi declines over time, other factors modulate viral reactivation in vivo despite robust HDAC activity. The identified host gene VOR biomarkers can be used for monitoring the pharmacodynamic activity of HDAC inhibitors. Histone deacetylase inhibitors (HDACi) are the most widely studied HIV latency-reversing agents (LRAs). The HDACi suberoylanilide hydroxamic acid (vorinostat [VOR]) has been employed in several clinical HIV latency reversal studies, as well as in vitro models of HIV latency, and has been shown to effectively induce HIV RNA and protein expression. Despite these findings, response to HDACi can vary, particularly with intermittent dosing, and information is lacking on the relationship between the host transcriptional response and HIV latency reversal. Here, we report on global gene expression responses to VOR and examine the longevity of the transcriptional response in various cellular models. We found that many genes are modulated at 6 h post-VOR treatment in HCT116, Jurkat, and primary resting CD4 T cells, yet return to baseline levels after an 18-h VOR-free period. With repeat exposure to VOR in resting CD4 T cells, we found similar and consistent transcriptional changes at 6 h following each serial treatment. In addition, serial exposure in HIV-infected suppressed donor CD4 T cells showed consistent transcriptional changes after each exposure to VOR. We identified five host genes that were strongly and consistently modulated following histone deacetylase (HDAC) inhibition; three (H1F0, IRGM, and WIPI49) were upregulated, and two (PHF15 and PRDM10) were downregulated. These genes demonstrated consistent modulation in peripheral blood mononuclear cell (PBMC) samples from HIV-positive (HIV+) participants who received either single or multiple doses of 400 mg of VOR. Interestingly, the host transcriptional response did not predict induction of cell-associated HIV RNA, suggesting that other cellular factors play key roles in HIV latency reversal in vivo despite robust HDACi pharmacological activity. IMPORTANCE Histone deacetylase inhibitors are widely studied HIV latency-reversing agents (LRAs). VOR, an HDACi, induces histone acetylation and chromatin remodeling and modulates host and HIV gene expression. However, the relationship between these events is poorly defined, and clinical studies suggest diminished HIV reactivation in resting CD4 T cells with daily exposure to VOR. Our study provides evidence that VOR induces a consistent level of host cell gene transcription following intermittent exposure. In addition, in response to VOR exposure a gene signature that was conserved across single and serial exposures both in vitro and in vivo was identified, indicating that VOR can consistently and reproducibly modulate transcriptional host responses. However, as the HIV response to HDACi declines over time, other factors modulate viral reactivation in vivo despite robust HDAC activity. The identified host gene VOR biomarkers can be used for monitoring the pharmacodynamic activity of HDAC inhibitors.
Collapse
|
184
|
Martinsen JT, Gunst JD, Højen JF, Tolstrup M, Søgaard OS. The Use of Toll-Like Receptor Agonists in HIV-1 Cure Strategies. Front Immunol 2020; 11:1112. [PMID: 32595636 PMCID: PMC7300204 DOI: 10.3389/fimmu.2020.01112] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/07/2020] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors and part of the first line of defense against invading microbes. In humans, we know of 10 different TLRs, which are expressed to varying degrees in immune cell subsets. Engaging TLRs through their specific ligands leads to activation of the innate immune system and secondarily priming of the adaptive immune system. Because of these unique properties, TLR agonists have been investigated as immunotherapy in cancer treatment for many years, but in recent years there has also been growing interest in the use of TLR agonists in the context of human immunodeficiency virus type 1 (HIV-1) cure research. The primary obstacle to curing HIV-1 is the presence of a latent viral reservoir in transcriptionally silent immune cells. Due to the very limited transcription of the integrated HIV-1 proviruses, latently infected cells cannot be targeted and cleared by immune effector mechanisms. TLR agonists are very interesting in this context because of their potential dual effects as latency reverting agents (LRAs) and immune modulatory compounds. Here, we review preclinical and clinical data on the impact of TLR stimulation on HIV-1 latency as well as antiviral and HIV-1-specific immunity. We also focus on the promising role of TLR agonists in combination strategies in HIV-1 cure research. Different combinations of TLR agonists and broadly neutralizing antibodies or TLRs agonists as adjuvants in HIV-1 vaccines have shown very encouraging results in non-human primate experiments and these concepts are now moving into clinical testing.
Collapse
Affiliation(s)
| | | | | | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
185
|
Abstract
Therapeutic approaches towards a functional cure or eradication of HIV have gained renewed momentum upon encouraging data emerging from studies in SIV monkey models and recent results from human clinical studies. However, a multitude of questions remain to be addressed, including how to deal with the latent viral reservoir, how to boost the host immune response to the virus and what the hurdles are to reach relevant viral compartments in the body. Advances have been made especially with regard to identifying agents that can reactivate the latent virus in vivo and boost the cellular and humoral immunity, but it remains largely unclear whether any of these strategies can awaken a sufficiently large fraction of the viral reservoir and whether the boosted immunity can prevent rapid viral replication once antiretroviral treatments are stopped.
Collapse
Affiliation(s)
- Lucia Bailon
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Beatriz Mothe
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain
| | | | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.
- Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.
- AELIX Therapeutics, Barcelona, Spain.
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
186
|
Mori L, Valente ST. Key Players in HIV-1 Transcriptional Regulation: Targets for a Functional Cure. Viruses 2020; 12:E529. [PMID: 32403278 PMCID: PMC7291152 DOI: 10.3390/v12050529] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
HIV-1 establishes a life-long infection when proviral DNA integrates into the host genome. The provirus can then either actively transcribe RNA or enter a latent state, without viral production. The switch between these two states is governed in great part by the viral protein, Tat, which promotes RNA transcript elongation. Latency is also influenced by the availability of host transcription factors, integration site, and the surrounding chromatin environment. The latent reservoir is established in the first few days of infection and serves as the source of viral rebound upon treatment interruption. Despite effective suppression of HIV-1 replication by antiretroviral therapy (ART), to below the detection limit, ART is ineffective at reducing the latent reservoir size. Elimination of this reservoir has become a major goal of the HIV-1 cure field. However, aside from the ideal total HIV-1 eradication from the host genome, an HIV-1 remission or functional cure is probably more realistic. The "block-and-lock" approach aims at the transcriptional silencing of the viral reservoir, to render suppressed HIV-1 promoters extremely difficult to reactivate from latency. There are unfortunately no clinically available HIV-1 specific transcriptional inhibitors. Understanding the mechanisms that regulate latency is expected to provide novel targets to be explored in cure approaches.
Collapse
Affiliation(s)
| | - Susana T. Valente
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA;
| |
Collapse
|
187
|
Mothe B, Rosás-Umbert M, Coll P, Manzardo C, Puertas MC, Morón-López S, Llano A, Miranda C, Cedeño S, López M, Alarcón-Soto Y, Melis GG, Langohr K, Barriocanal AM, Toro J, Ruiz I, Rovira C, Carrillo A, Meulbroek M, Crook A, Wee EG, Miró JM, Clotet B, Valle M, Martinez-Picado J, Hanke T, Brander C, Moltó J. HIVconsv Vaccines and Romidepsin in Early-Treated HIV-1-Infected Individuals: Safety, Immunogenicity and Effect on the Viral Reservoir (Study BCN02). Front Immunol 2020; 11:823. [PMID: 32435247 PMCID: PMC7218169 DOI: 10.3389/fimmu.2020.00823] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/09/2020] [Indexed: 12/30/2022] Open
Abstract
Kick&kill strategies combining drugs aiming to reactivate the viral reservoir with therapeutic vaccines to induce effective cytotoxic immune responses hold potential to achieve a functional cure for HIV-1 infection. Here, we report on an open-label, single-arm, phase I clinical trial, enrolling 15 early-treated HIV-1-infected individuals, testing the combination of the histone deacetylase inhibitor romidepsin as a latency-reversing agent and the MVA.HIVconsv vaccine. Romidepsin treatment resulted in increased histone acetylation, cell-associated HIV-1 RNA, and T-cell activation, which were associated with a marginally significant reduction of the viral reservoir. Vaccinations boosted robust and broad HIVconsv-specific T cells, which were strongly refocused toward conserved regions of the HIV-1 proteome. During a monitored ART interruption phase using plasma viral load over 2,000 copies/ml as a criterium for ART resumption, 23% of individuals showed sustained suppression of viremia up to 32 weeks without evidence for reseeding the viral reservoir. Results from this pilot study show that the combined kick&kill intervention was safe and suggest a role for this strategy in achieving an immune-driven durable viremic control.
Collapse
Affiliation(s)
- Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain
| | - Miriam Rosás-Umbert
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Pep Coll
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
| | | | | | | | - Anuska Llano
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
| | - Cristina Miranda
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Miriam López
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Yovaninna Alarcón-Soto
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Guadalupe Gómez Melis
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Klaus Langohr
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya/BARCELONATECH, Barcelona, Spain
| | - Ana M Barriocanal
- Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain.,Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Jessica Toro
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Irene Ruiz
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Cristina Rovira
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Antonio Carrillo
- Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | | | - Alison Crook
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Edmund G Wee
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jose M Miró
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Marta Valle
- Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona (UAB), Barcelona, Spain.,Pharmacokinetic/Pharmacodynamic Modeling and Simultation, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,ICREA, Barcelona, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, United Kingdom.,Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,ICREA, Barcelona, Spain
| | - José Moltó
- Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Infectious Diseases, Germans Trias i Pujol Research Institute, Badalona, Spain
| | | |
Collapse
|
188
|
Prodrugs of PKC modulators show enhanced HIV latency reversal and an expanded therapeutic window. Proc Natl Acad Sci U S A 2020; 117:10688-10698. [PMID: 32371485 DOI: 10.1073/pnas.1919408117] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIDS is a pandemic disease caused by HIV that affects 37 million people worldwide. Current antiretroviral therapy slows disease progression but does not eliminate latently infected cells, which resupply active virus, thus necessitating lifelong treatment with associated compliance, cost, and chemoexposure issues. Latency-reversing agents (LRAs) activate these cells, allowing for their potential clearance, thus presenting a strategy to eradicate the infection. Protein kinase C (PKC) modulators-including prostratin, ingenol esters, bryostatin, and their analogs-are potent LRAs in various stages of development for several clinical indications. While LRAs are promising, a major challenge associated with their clinical use is sustaining therapeutically meaningful levels of the active agent while minimizing side effects. Here we describe a strategy to address this problem based on LRA prodrugs, designed for controllable release of the active LRA after a single injection. As intended, these prodrugs exhibit comparable or superior in vitro activity relative to the parent compounds. Selected compounds induced higher in vivo expression of CD69, an activation biomarker, and, by releasing free agent over time, significantly improved tolerability when compared to the parent LRAs. More generally, selected prodrugs of PKC modulators avoid the bolus toxicities of the parent drug and exhibit greater efficacy and expanded tolerability, thereby addressing a longstanding objective for many clinical applications.
Collapse
|
189
|
PIWIL4 Maintains HIV-1 Latency by Enforcing Epigenetically Suppressive Modifications on the 5' Long Terminal Repeat. J Virol 2020; 94:JVI.01923-19. [PMID: 32161174 DOI: 10.1128/jvi.01923-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Although substantial progress has been made in depicting the molecular pathogenesis of human immunodeficiency virus type 1 (HIV-1) infection, the comprehensive mechanism of HIV-1 latency and the most promising therapeutic strategies to effectively reactivate the HIV-1 latent reservoir to achieve a functional cure for AIDS remain to be systematically illuminated. Here, we demonstrated that piwi (P element-induced Wimpy)-like RNA-mediated gene silencing 4 (PIWIL4) played an important role in suppressing HIV-1 transcription and contributed to the latency state in HIV-1-infected cells through its recruitment of various suppressive factors, including heterochromatin protein 1α/β/γ, SETDB1, and HDAC4. The knockdown of PIWIL4 enhanced HIV-1 transcription and reversed HIV-1 latency in both HIV-1 latently infected Jurkat T cells and primary CD4+ T lymphocytes and resting CD4+ T lymphocytes from HIV-1-infected individuals on suppressive combined antiretroviral therapy (cART). Furthermore, in the absence of PIWIL4, HIV-1 latently infected Jurkat T cells were more sensitive to reactivation with vorinostat (suberoylanilide hydroxamic acid, or SAHA), JQ1, or prostratin. These findings indicated that PIWIL4 promotes HIV-1 latency by imposing repressive marks at the HIV-1 5' long terminal repeat. Thus, the manipulation of PIWIL4 could be a novel strategy for developing promising latency-reversing agents (LRAs).IMPORTANCE HIV-1 latency is systematically modulated by host factors and viral proteins. During this process, the suppression of HIV-1 transcription plays an essential role in promoting HIV-1 latency. In this study, we found that PIWIL4 repressed HIV-1 promoter activity and maintained HIV-1 latency. In particular, we report that PIWIL4 can regulate gene expression through its association with the suppressive activity of HDAC4. Therefore, we have identified a new function for PIWIL4: it is not only a suppressor of endogenous retrotransposons but also plays an important role in inhibiting transcription and leading to latent infection of HIV-1, a well-known exogenous retrovirus. Our results also indicate a novel therapeutic target to reactivate the HIV-1 latent reservoir.
Collapse
|
190
|
Hashemi P, Sadowski I. Diversity of small molecule HIV-1 latency reversing agents identified in low- and high-throughput small molecule screens. Med Res Rev 2020; 40:881-908. [PMID: 31608481 PMCID: PMC7216841 DOI: 10.1002/med.21638] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
The latency phenomenon produced by human immunodeficiency virus (HIV-1) prevents viral clearance by current therapies, and consequently development of a cure for HIV-1 disease represents a formidable challenge. Research over the past decade has resulted in identification of small molecules that are capable of exposing HIV-1 latent reservoirs, by reactivation of viral transcription, which is intended to render these infected cells sensitive to elimination by immune defense recognition or apoptosis. Molecules with this capability, known as latency-reversing agents (LRAs) could lead to realization of proposed HIV-1 cure strategies collectively termed "shock and kill," which are intended to eliminate the latently infected population by forced reactivation of virus replication in combination with additional interventions that enhance killing by the immune system or virus-mediated apoptosis. Here, we review efforts to discover novel LRAs via low- and high-throughput small molecule screens, and summarize characteristics and biochemical properties of chemical structures with this activity. We expect this analysis will provide insight toward further research into optimized designs for new classes of more potent LRAs.
Collapse
Affiliation(s)
- Pargol Hashemi
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ivan Sadowski
- Biochemistry and Molecular Biology, Molecular Epigenetics, Life Sciences InstituteUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
191
|
Chen L, Quan H, Xu Z, Wang H, Xia Y, Lou L, Yang W. A modular biomimetic strategy for the synthesis of macrolide P-glycoprotein inhibitors via Rh-catalyzed C-H activation. Nat Commun 2020; 11:2151. [PMID: 32358512 PMCID: PMC7195407 DOI: 10.1038/s41467-020-16084-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/09/2020] [Indexed: 11/09/2022] Open
Abstract
One of the key challenges to overcome multidrug resistance (MDR) in cancer is the development of more effective and general strategies to discover bioactive scaffolds. Inspired by natural products, we describe a strategy to achieve this goal by modular biomimetic synthesis of scaffolds of (Z)-allylic-supported macrolides. Herein, an Rh(III)-catalyzed native carboxylic acid-directed and solvent-free C−H activation allylation with high stereoselectivity and chemoselectivity is achieved. The generated poly-substituted allylic alcohol as a multifunctional and biomimetic building block is crucial for the synthesis of (Z)-allylic-supported macrolides. Moreover, the unique allylic-supported macrolides significantly potentiate the sensitivity of tumor cells to cytotoxic agents such as vinorelbine and doxetaxel by reversing p170-glycoprotein-mediated MDR. Our findings will inspire the evolution of synthetic chemistry and open avenues for expedient and diversified synthesis of bioactive macrocyclic molecules. One strategy to address multidrug resistance in cancer is the development of modular methods to access bioactive scaffolds. Here, the authors report a Rh(III)-catalyzed carboxylic acid-directed C(sp2)−H allylation and apply it to the modular synthesis of (Z)-allylic macrolides which enhance antitumoral drug activity.
Collapse
Affiliation(s)
- Lu Chen
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haitian Quan
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongliang Xu
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Wang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanzhi Xia
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325035, China
| | - Liguang Lou
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Weibo Yang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Key Laboratory for Functional Material, Educational Department of Liaoning Province, University of Science and Technology Liaoning, Anshan, 114051, China.
| |
Collapse
|
192
|
Ren Y, Huang SH, Patel S, Alberto WDC, Magat D, Ahimovic D, Macedo AB, Durga R, Chan D, Zale E, Mota TM, Truong R, Rohwetter T, McCann CD, Kovacs CM, Benko E, Wimpelberg A, Cannon C, Hardy WD, Bosque A, Bollard CM, Jones RB. BCL-2 antagonism sensitizes cytotoxic T cell-resistant HIV reservoirs to elimination ex vivo. J Clin Invest 2020; 130:2542-2559. [PMID: 32027622 PMCID: PMC7191002 DOI: 10.1172/jci132374] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/29/2020] [Indexed: 12/11/2022] Open
Abstract
Curing HIV infection will require the elimination of a reservoir of infected CD4+ T cells that persists despite HIV-specific cytotoxic T cell (CTL) responses. Although viral latency is a critical factor in this persistence, recent evidence also suggests a role for intrinsic resistance of reservoir-harboring cells to CTL killing. This resistance may have contributed to negative outcomes of clinical trials, where pharmacologic latency reversal has thus far failed to drive reductions in HIV reservoirs. Through transcriptional profiling, we herein identified overexpression of the prosurvival factor B cell lymphoma 2 (BCL-2) as a distinguishing feature of CD4+ T cells that survived CTL killing. We show that the inducible HIV reservoir was disproportionately present in BCL-2hi subsets in ex vivo CD4+ T cells. Treatment with the BCL-2 antagonist ABT-199 was not sufficient to drive reductions in ex vivo viral reservoirs when tested either alone or with a latency-reversing agent (LRA). However, the triple combination of strong LRAs, HIV-specific T cells, and a BCL-2 antagonist uniquely enabled the depletion of ex vivo viral reservoirs. Our results provide rationale for novel therapeutic approaches targeting HIV cure and, more generally, suggest consideration of BCL-2 antagonism as a means of enhancing CTL immunotherapy in other settings, such as cancer.
Collapse
Affiliation(s)
- Yanqin Ren
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Szu Han Huang
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Shabnum Patel
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC, USA
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Winiffer D. Conce Alberto
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Dean Magat
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Dughan Ahimovic
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Amanda B. Macedo
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Ryan Durga
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Dora Chan
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Elizabeth Zale
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Talia M. Mota
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Ronald Truong
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Thomas Rohwetter
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Chase D. McCann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Erika Benko
- Maple Leaf Medical Clinic, Toronto, Ontario, Canada
| | | | | | - W. David Hardy
- Whitman-Walker Health, Washington, DC, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alberto Bosque
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - Catherine M. Bollard
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC, USA
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| | - R. Brad Jones
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, DC, USA
| |
Collapse
|
193
|
Mota TM, McCann CD, Danesh A, Huang SH, Magat DB, Ren Y, Leyre L, Bui TD, Rohwetter TM, Kovacs CM, Benko E, MacLaren L, Wimpelberg A, Cannon CM, Hardy WD, Safrit JT, Jones RB. Integrated Assessment of Viral Transcription, Antigen Presentation, and CD8 + T Cell Function Reveals Multiple Limitations of Class I-Selective Histone Deacetylase Inhibitors during HIV-1 Latency Reversal. J Virol 2020; 94:e01845-19. [PMID: 32051267 PMCID: PMC7163115 DOI: 10.1128/jvi.01845-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022] Open
Abstract
Clinical trials investigating histone deacetylase inhibitors (HDACi) to reverse HIV-1 latency aim to expose reservoirs in antiretroviral (ARV)-treated individuals to clearance by immune effectors, yet have not driven measurable reductions in the frequencies of infected cells. We therefore investigated the effects of the class I-selective HDACi nanatinostat and romidepsin on various blocks to latency reversal and elimination, including viral splicing, antigen presentation, and CD8+ T cell function. In ex vivo CD4+ T cells from ARV-suppressed individuals, both HDACi significantly induced viral transcription, but not splicing nor supernatant HIV-1 RNA. In an HIV-1 latency model using autologous CD8+ T cell clones as biosensors of antigen presentation, neither HDACi-treated CD4+ T cell condition induced clone degranulation. Both HDACi also impaired the function of primary CD8+ T cells in viral inhibition assays, with nanatinostat causing less impairment. These findings suggest that spliced or cell-free HIV-1 RNAs are more indicative of antigen expression than unspliced HIV-RNAs and may help to explain the limited abilities of HDACi to generate CD8+ T cell targets in vivoIMPORTANCE Antiretroviral (ARV) drug regimens suppress HIV-1 replication but are unable to cure infection. This leaves people living with HIV-1 burdened by a lifelong commitment to expensive daily medication. Furthermore, it has become clear that ARV therapy does not fully restore health, leaving individuals at elevated risk for cardiovascular disease, certain types of cancers, and neurocognitive disorders, as well as leaving them exposed to stigma. Efforts are therefore under way to develop therapies capable of curing infection. A key focus of these efforts has been on a class of drugs called histone deacetylase inhibitors (HDACi), which have the potential of exposing hidden reservoirs of HIV-1 to elimination by the immune system. Unfortunately, clinical trial results with HDACi have thus far been disappointing. In the current study, we integrate a number of experimental approaches to build a model that provides insights into the limited activity of HDACi in clinical trials and offers direction for future approaches.
Collapse
Affiliation(s)
- Talia M Mota
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Chase D McCann
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Ali Danesh
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Szu-Han Huang
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Dean B Magat
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Yanqin Ren
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Louise Leyre
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Tracy D Bui
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Thomas M Rohwetter
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | | | | | - Lynsay MacLaren
- Research Department, Whitman-Walker Health, Washington, DC, USA
| | | | | | - W David Hardy
- Division of Infectious Disease, Johns Hopkins University School of Medicine, Washington, DC, USA
| | | | - R Brad Jones
- Infectious Diseases Division, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
- Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
194
|
Margolis DM, Archin NM, Cohen MS, Eron JJ, Ferrari G, Garcia JV, Gay CL, Goonetilleke N, Joseph SB, Swanstrom R, Turner AMW, Wahl A. Curing HIV: Seeking to Target and Clear Persistent Infection. Cell 2020; 181:189-206. [PMID: 32220311 PMCID: PMC7896558 DOI: 10.1016/j.cell.2020.03.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection persists despite years of antiretroviral therapy (ART). To remove the stigma and burden of chronic infection, approaches to eradicate or cure HIV infection are desired. Attempts to augment ART with therapies that reverse viral latency, paired with immunotherapies to clear infection, have advanced into the clinic, but the field is still in its infancy. We review foundational studies and highlight new insights in HIV cure research. Together with advances in ART delivery and HIV prevention strategies, future therapies that clear HIV infection may relieve society of the affliction of the HIV pandemic.
Collapse
Affiliation(s)
- David M Margolis
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC 27599, USA.
| | - Nancie M Archin
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Myron S Cohen
- Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joseph J Eron
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Guido Ferrari
- Department of Surgery and Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - J Victor Garcia
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Cynthia L Gay
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Nilu Goonetilleke
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah B Joseph
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ronald Swanstrom
- Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Anne-Marie W Turner
- UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Angela Wahl
- International Center for the Advancement of Translational Science, Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA; Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
195
|
High levels of genetically intact HIV in HLA-DR+ memory T cells indicates their value for reservoir studies. AIDS 2020; 34:659-668. [PMID: 31913161 DOI: 10.1097/qad.0000000000002465] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The contribution of HLA-DR+ memory CD4 T cells to the HIV reservoir during prolonged antiretroviral therapy is unclear as these cells are commonly excluded when assessing for replication-competent HIV. To address this issue, we examined the distribution of genetically intact HIV DNA within HLA-DR- and HLA-DR+ memory CD4 T cells and the RNA transcriptional profile of these cells during antiretroviral therapy. DESIGN/METHODS Full-length DNA sequencing was used to examine the HIV DNA landscape within HLA-DR+ and HLA-DR- memory CD4 T cells. RNA quantification and sequencing was used to interrogate the relationship between HLA-DR status and HIV RNA transcription. RESULTS HLA-DR+ CD4 T cells contained a high frequency of genetically intact HIV genomes, contributing over half of the genetically intact viral sequences to the reservoir. Expansions of genetically identical sequences were identified in all T-cell subsets, indicating that cellular proliferation maintains genetically intact and defective viral DNA during therapy. Intracellular HIV RNA levels in HLA-DR+ and HLA-DR- T cells were not statistically different by either long terminal repeat quantitative PCR quantification or single-genome RNA sequencing of the p6-RT region. CONCLUSION The high proportion of intact viral DNA sequences in the proliferative HLA-DR+ subset suggests they are critical in maintaining HIV infection during effective therapy. As such, these cells should be included in any immune intervention targeting HIV during effective therapy.
Collapse
|
196
|
De la Torre-Tarazona HE, Jiménez R, Bueno P, Camarero S, Román L, Fernández-García JL, Beltrán M, Nothias LF, Cachet X, Paolini J, Litaudon M, Alcami J, Bedoya LM. 4-Deoxyphorbol inhibits HIV-1 infection in synergism with antiretroviral drugs and reactivates viral reservoirs through PKC/MEK activation synergizing with vorinostat. Biochem Pharmacol 2020; 177:113937. [PMID: 32224142 DOI: 10.1016/j.bcp.2020.113937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/24/2020] [Indexed: 01/06/2023]
Abstract
Latent HIV reservoirs are the main obstacle to eradicate HIV infection. One strategy proposes to eliminate these viral reservoirs by pharmacologically reactivating the latently infected T cells. We show here that a 4-deoxyphorbol ester derivative isolated from Euphorbia amygdaloides ssp. semiperfoliata, 4β-dPE A, reactivates HIV-1 from latency and could potentially contribute to decrease the viral reservoir. 4β-dPE A shows two effects in the HIV replication cycle, infection inhibition and HIV transactivation, similarly to other phorboids PKC agonists such PMA and prostratin and to other diterpene esters such SJ23B. Our data suggest 4β-dPE A is non-tumorigenic, unlike the related compound PMA. As the compounds are highly similar, the lack of tumorigenicity by 4β-dPE A could be due to the lack of a long side lipophilic chain that is present in PMA. 4β-dPE activates HIV transcription at nanomolar concentrations, lower than the concentration needed by other latency reversing agents (LRAs) such as prostratin and similar to bryostatin. PKCθ/MEK activation is required for the transcriptional activity, and thus, anti-latency activity of 4β-dPE A. However, CD4, CXCR4 and CCR5 receptors down-regulation effect seems to be independent of PCK/MEK, suggesting the existence of at least two different targets for 4β-dPE A. Furthermore, NF-κb transcription factor is involved in 4β-dPE HIV reactivation, as previously shown for other PKCs agonists. We also studied the effects of 4β-dPE A in combination with other LRAs. When 4β-dPE A was combined with another PKC agonists such as prostratin an antagonic effect was achieved, while, when combined with an HDAC inhibitor such as vorinostat, a strong synergistic effect was obtained. Interestingly, the latency reversing effect of the combination was synergistically diminishing the EC50 value but also increasing the efficacy showed by the drugs alone. In addition, combinations of 4β-dPE A with antiretroviral drugs as CCR5 antagonist, NRTIs, NNRTIs and PIs, showed a consistent synergistic effect, suggesting that the combination would not interefer with antiretroviral therapy (ART). Finally, 4β-dPE A induced latent HIV reactivation in CD4 + T cells of infected patients under ART at similar levels than the tumorigenic phorbol derivative PMA, showing a clear reactivation effect. In summary, we describe here the mechanism of action of a new potent deoxyphorbol derivative as a latency reversing agent candidate to decrease the size of HIV reservoirs.
Collapse
Affiliation(s)
- H E De la Torre-Tarazona
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - R Jiménez
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - P Bueno
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - S Camarero
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - L Román
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - J L Fernández-García
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain; Pharmacology Department, Pharmacy Faculty, Universidad Complutense de Madrid, Pz. Ramón Y Cajal s/n, 28040 Madrid, Spain
| | - M Beltrán
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain
| | - L F Nothias
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, University of Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - X Cachet
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, University of Paris-Saclay, 91198 Gif-sur-Yvette, France; CiTCoM Laboratory, UMR 8038 CNRS-University of Paris, Faculty of Pharmacy, University of Paris, 75006 Paris, France
| | - J Paolini
- Laboratoire de Chimie des Produits Naturels, CNRS, UMR SPE 6134, University of Corsica, 20250 Corte, France
| | - M Litaudon
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, University of Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - J Alcami
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain; Infectious Diseases Unit, IBIDAPS, Hospital Clínic, University of Barcelona, Spain.
| | - L M Bedoya
- AIDS Immunopathology Department, National Centre of Microbiology, Instituto de Salud Carlos III. Ctra. Pozuelo Km. 2. Majadahonda, 28224 Madrid, Spain; Pharmacology Department, Pharmacy Faculty, Universidad Complutense de Madrid, Pz. Ramón Y Cajal s/n, 28040 Madrid, Spain.
| |
Collapse
|
197
|
CRISPR-based gene knockout screens reveal deubiquitinases involved in HIV-1 latency in two Jurkat cell models. Sci Rep 2020; 10:5350. [PMID: 32210344 PMCID: PMC7093534 DOI: 10.1038/s41598-020-62375-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
The major barrier to a HIV-1 cure is the persistence of latent genomes despite treatment with antiretrovirals. To investigate host factors which promote HIV-1 latency, we conducted a genome-wide functional knockout screen using CRISPR-Cas9 in a HIV-1 latency cell line model. This screen identified IWS1, POLE3, POLR1B, PSMD1, and TGM2 as potential regulators of HIV-1 latency, of which PSMD1 and TMG2 could be confirmed pharmacologically. Further investigation of PSMD1 revealed that an interacting enzyme, the deubiquitinase UCH37, was also involved in HIV-1 latency. We therefore conducted a comprehensive evaluation of the deubiquitinase family by gene knockout, identifying several deubiquitinases, UCH37, USP14, OTULIN, and USP5 as possible HIV-1 latency regulators. A specific inhibitor of USP14, IU1, reversed HIV-1 latency and displayed synergistic effects with other latency reversal agents. IU1 caused degradation of TDP-43, a negative regulator of HIV-1 transcription. Collectively, this study is the first comprehensive evaluation of deubiquitinases in HIV-1 latency and establishes that they may hold a critical role.
Collapse
|
198
|
Assessing the impact of AGS-004, a dendritic cell-based immunotherapy, and vorinostat on persistent HIV-1 Infection. Sci Rep 2020; 10:5134. [PMID: 32198428 PMCID: PMC7083965 DOI: 10.1038/s41598-020-61878-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/03/2020] [Indexed: 11/09/2022] Open
Abstract
Approaches to deplete persistent HIV infection are needed. We investigated the combined impact of the latency reversing agent vorinostat (VOR) and AGS-004, an autologous dendritic cell immunotherapeutic, on the HIV reservoir. HIV+, stably treated participants in whom resting CD4+ T cell-associated HIV RNA (rca-RNA) increased after VOR exposure ex vivo and in vivo received 4 doses of AGS-004 every 3 weeks, followed by VOR every 72 hours for 30 days, and then the cycle repeated. Change in VOR-responsive host gene expression, HIV-specific T cell responses, low-level HIV viremia, rca-RNA, and the frequency of resting CD4+ T-cell infection (RCI) was measured at baseline and after each cycle. No serious treatment-related adverse events were observed among five participants. As predicted, VOR-responsive host genes responded uniformly to VOR dosing. Following cycles of AGS-004 and VOR, rca-RNA decreased significantly in only two participants, with a significant decrease in SCA observed in one of these participants. However, unlike other cohorts dosed with AGS-004, no uniform increase in HIV-specific immune responses following vaccination was observed. Finally, no reproducible decline of RCI, defined as a decrease of >50%, was observed. AGS-004 and VOR were safe and well-tolerated, but no substantial impact on RCI was measured. In contrast to previous clinical data, AGS-004 did not induce HIV-specific immune responses greater than those measured at baseline. More efficacious antiviral immune interventions, perhaps paired with more effective latency reversal, must be developed to clear persistent HIV infection.
Collapse
|
199
|
Rosás-Umbert M, Ruiz-Riol M, Fernández MA, Marszalek M, Coll P, Manzardo C, Cedeño S, Miró JM, Clotet B, Hanke T, Moltó J, Mothe B, Brander C. In vivo Effects of Romidepsin on T-Cell Activation, Apoptosis and Function in the BCN02 HIV-1 Kick&Kill Clinical Trial. Front Immunol 2020; 11:418. [PMID: 32265913 PMCID: PMC7100631 DOI: 10.3389/fimmu.2020.00418] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/24/2020] [Indexed: 12/17/2022] Open
Abstract
Romidepsin (RMD) is a well-characterized histone deacetylase inhibitor approved for the treatment of cutaneous T-cell lymphoma. in vitro and in vivo studies have demonstrated that it is able to induce HIV-1 gene expression in latently infected CD4+ T cells from HIV-1+ individuals on suppressive antiretroviral therapy. However, in vitro experiments suggested that RMD could also impair T-cell functionality, particularly of activated T cells. Thus, the usefulness of RMD in HIV-1 kick&kill strategies, that aim to enhance the immune system elimination of infected cells after inducing HIV-1 viral reactivation, may be limited. In order to address whether the in vitro observations are replicated in vivo, we determined the effects of RMD on the total and HIV-1-specific T-cell populations in longitudinal samples from the BCN02 kick&kill clinical trial (NCT02616874). BCN02 was a proof-of-concept study in 15 early treated HIV-1+ individuals that combined MVA.HIVconsv vaccination with three weekly infusions of RMD given as a latency reversing agent. Our results show that RMD induced a transient increase in the frequency of apoptotic T cells and an enhanced activation of vaccine-induced T cells. Although RMD reduced the number of vaccine-elicited T cells secreting multiple cytokines, viral suppressive capacity of CD8+ T cells was preserved over the RMD treatment. These observations have important implications for the design of effective kick&kill strategies for the HIV-1 cure.
Collapse
Affiliation(s)
- Miriam Rosás-Umbert
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | | | - Marco A Fernández
- Flow Cytometry Facility, Health Sciences Research Institute Germans Trias i Pujol, Badalona, Spain
| | | | - Pep Coll
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
| | | | | | - José M Miró
- Hospital Clinic- IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Spain.,Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic - Central University of Catalonia (UVic - UCC), Vic, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford, United Kingdom.,Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - José Moltó
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Department of Infectious Diseases, Hospital Germans Trias i Pujol, Badalona, Spain.,Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic - Central University of Catalonia (UVic - UCC), Vic, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain.,Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic - Central University of Catalonia (UVic - UCC), Vic, Spain.,ICREA, Pg. Lluis Companys, Barcelona, Spain
| | | |
Collapse
|
200
|
Boucau J, Das J, Joshi N, Le Gall S. Latency reversal agents modulate HIV antigen processing and presentation to CD8 T cells. PLoS Pathog 2020; 16:e1008442. [PMID: 32196533 PMCID: PMC7112239 DOI: 10.1371/journal.ppat.1008442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 04/01/2020] [Accepted: 02/28/2020] [Indexed: 01/03/2023] Open
Abstract
Latency reversal agents (LRA) variably induce HIV re-expression in CD4 T cells but reservoirs are not cleared. Whether HIV epitope presentation is similar between latency reversal and initial infection of CD4 T cells is unknown yet crucial to define immune responses able to detect HIV-infected CD4 T cells after latency reversal. HIV peptides displayed by MHC comes from the intracellular degradation of proteins by proteasomes and post-proteasomal peptidases but the impact of LRAs on antigen processing is not known. Here we show that HDAC inhibitors (HDCAi) reduced cytosolic proteolytic activities while PKC agonists (PKCa) increased them to a lesser extent than that induced by TCR activation. During the cytosolic degradation of long HIV peptides in LRA-treated CD4 T cells extracts, HDACi and PKCa modulated degradation patterns of peptides and altered the production of HIV epitopes in often opposite ways. Beyond known HIV epitopes, HDACi narrowed the coverage of HIV antigenic fragments by 8-11aa degradation peptides while PKCa broadened it. LRAs altered HIV infection kinetics and modulated CD8 T cell activation in an epitope- and time-dependent manner. Interestingly the efficiency of endogenous epitope processing and presentation to CD8 T cells was increased by PKCa Ingenol at early time points despite low levels of antigens. LRA-induced modulations of antigen processing should be considered and exploited to enhance and broaden HIV peptide presentation by CD4 T cells and to improve immune recognition after latency reversal. This property of LRAs, if confirmed with other antigens, might be exploited to improve immune detection of diseased cells beyond HIV. Latently HIV-infected CD4 T cells persist and remain invisible to the immune system. Strategies to flush out HIV reservoirs propose to re-express HIV with latency reversal agents (LRAs), leading to CD4 T cell death or clearance by HIV-specific immune responses. LRAs tested so far variably induced HIV re-expression but did not eliminate reservoirs. The activation of HIV-specific immune responses is triggered by HIV peptides displayed by infected cells after HIV intracellular degradation. Whether HIV antigens are similarly degraded and displayed by CD4 T cells after latency reversal or during initial infection is unknown. We showed that LRAs altered the activities of the degradation machinery and changed the degradation patterns of HIV into peptides. LRA-treated HIV-infected CD4 T cells were variably recognized by immune cells in a time- and peptide-dependent manner. Some LRAs increased the efficiency of HIV peptide presentation despite low levels of HIV antigens inside CD4 T cells. The modulation of HIV peptide presentation by current or future LRAs should be accounted for and exploited to improve HIV peptide presentation and enhance immune detection of HIV-infected CD4 T cells after latency reversal.
Collapse
Affiliation(s)
- Julie Boucau
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Jishnu Das
- Center for Systems Immunology, Departments of Immunology and Computational & Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Neelambari Joshi
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Sylvie Le Gall
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|