Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
STAR Protoc ; 4(2): 102253, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37097819

ABSTRACT

Integrated HIV-1 DNA persists in cells of people living with HIV during antiretroviral treatment, but its quantification is hindered by its rarity. Here, we present an optimized protocol to evaluate "shock and kill" therapeutic strategies, including both the latency reactivation ("shock") and elimination of infected cells ("kill") stages. We describe steps for the sequential use of nested PCR-based assays and viability sorting to allow for scalable and rapid screening of candidate therapeutics in patient-derived blood cells. For complete details on the use and execution of this protocol, please refer to Shytaj et al..1.

2.
mBio ; 13(2): e0370521, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35229634

ABSTRACT

Combinations of direct-acting antivirals are needed to minimize drug resistance mutations and stably suppress replication of RNA viruses. Currently, there are limited therapeutic options against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and testing of a number of drug regimens has led to conflicting results. Here, we show that cobicistat, which is an FDA-approved drug booster that blocks the activity of the drug-metabolizing proteins cytochrome P450-3As (CYP3As) and P-glycoprotein (P-gp), inhibits SARS-CoV-2 replication. Two independent cell-to-cell membrane fusion assays showed that the antiviral effect of cobicistat is exerted through inhibition of spike protein-mediated membrane fusion. In line with this, incubation with low-micromolar concentrations of cobicistat decreased viral replication in three different cell lines including cells of lung and gut origin. When cobicistat was used in combination with remdesivir, a synergistic effect on the inhibition of viral replication was observed in cell lines and in a primary human colon organoid. This was consistent with the effects of cobicistat on two of its known targets, CYP3A4 and P-gp, the silencing of which boosted the in vitro antiviral activity of remdesivir in a cobicistat-like manner. When administered in vivo to Syrian hamsters at a high dose, cobicistat decreased viral load and mitigated clinical progression. These data highlight cobicistat as a therapeutic candidate for treating SARS-CoV-2 infection and as a potential building block of combination therapies for COVID-19. IMPORTANCE The lack of effective antiviral treatments against SARS-CoV-2 is a significant limitation in the fight against the COVID-19 pandemic. Single-drug regimens have so far yielded limited results, indicating that combinations of antivirals might be required, as previously seen for other RNA viruses. Our work introduces the drug booster cobicistat, which is approved by the FDA and typically used to potentiate the effect of anti-HIV protease inhibitors, as a candidate inhibitor of SARS-CoV-2 replication. Beyond its direct activity as an antiviral, we show that cobicistat can enhance the effect of remdesivir, which was one of the first drugs proposed for treatment of SARS-CoV-2. Overall, the dual action of cobicistat as a direct antiviral and a drug booster can provide a new approach to design combination therapies and rescue the activity of compounds that are only partially effective in monotherapy.


Subject(s)
COVID-19 Drug Treatment , Hepatitis C, Chronic , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cobicistat , Cricetinae , Disease Progression , Humans , Mesocricetus , Pandemics , SARS-CoV-2 , Viral Load
3.
AIDS Res Ther ; 19(1): 2, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35022035

ABSTRACT

BACKGROUND: We developed a personalized Monocyte-Derived Dendritic-cell Therapy (MDDCT) for HIV-infected individuals on suppressive antiretroviral treatment and evaluated HIV-specific T-cell responses. METHODS: PBMCs were obtained from 10 HIV+ individuals enrolled in trial NCT02961829. Monocytes were differentiated into DCs using IFN-α and GM-CSF. After sequencing each patient's HIV-1 Gag and determining HLA profiles, autologous Gag peptides were selected based on the predicted individual immunogenicity and used to pulse MDDCs. Three doses of the MDDCT were administered every 15 days. To assess immunogenicity, patients' cells were stimulated in vitro with autologous peptides, and intracellular IL-2, TNF, and interferon-gamma (IFN-γ) production were measured in CD4+ and CD8+ T-cells. RESULTS: The protocol of ex-vivo treatment with IFN-α and GM-CSF was able to induce maturation of MDDCs, as well as to preserve their viability for reinfusion. MDDCT administration was associated with increased expression of IL-2 in CD4+ and CD8+ T-cells at 15 and/or 30 days after the first MDDCT administration. Moreover, intracellular TNF and IFN-γ expression was significantly increased in CD4+ T-cells. The number of candidates that increased in vitro the cytokine levels in CD4+ and CD8+ T cells upon stimulation with Gag peptides from baseline to day 15 and from baseline to day 30 and day 120 after MDDCT was significant as compared to Gag unstimulated response. This was accompanied by an increasing trend in the frequency of polyfunctional T-cells over time, which was visible when considering both cells expressing two and three out of the three cytokines examined. CONCLUSIONS: MDDC had a mature profile, and this MDDCT promoted in-vitro T-cell immune responses in HIV-infected patients undergoing long-term suppressive antiretroviral treatment. Trial registration NCT02961829: (Multi Interventional Study Exploring HIV-1 Residual Replication: a Step Towards HIV-1 Eradication and Sterilizing Cure, https://www.clinicaltrials.gov/ct2/show/NCT02961829 , posted November 11th, 2016).


Subject(s)
HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cell- and Tissue-Based Therapy , Dendritic Cells , HIV Infections/drug therapy , Humans
4.
EMBO Mol Med ; 13(8): e13901, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34289240

ABSTRACT

HIV-1 infects lymphoid and myeloid cells, which can harbor a latent proviral reservoir responsible for maintaining lifelong infection. Glycolytic metabolism has been identified as a determinant of susceptibility to HIV-1 infection, but its role in the development and maintenance of HIV-1 latency has not been elucidated. By combining transcriptomic, proteomic, and metabolomic analyses, we here show that transition to latent HIV-1 infection downregulates glycolysis, while viral reactivation by conventional stimuli reverts this effect. Decreased glycolytic output in latently infected cells is associated with downregulation of NAD+ /NADH. Consequently, infected cells rely on the parallel pentose phosphate pathway and its main product, NADPH, fueling antioxidant pathways maintaining HIV-1 latency. Of note, blocking NADPH downstream effectors, thioredoxin and glutathione, favors HIV-1 reactivation from latency in lymphoid and myeloid cellular models. This provides a "shock and kill effect" decreasing proviral DNA in cells from people living with HIV/AIDS. Overall, our data show that downmodulation of glycolysis is a metabolic signature of HIV-1 latency that can be exploited to target latently infected cells with eradication strategies.


Subject(s)
HIV Infections , HIV-1 , CD4-Positive T-Lymphocytes , Down-Regulation , Glycolysis , Humans , Oxidative Stress , Proteomics , Virus Activation , Virus Latency
5.
Eur J Drug Metab Pharmacokinet ; 45(6): 715-723, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32780273

ABSTRACT

BACKGROUND AND OBJECTIVES: Chloroquine/hydroxychloroquine has recently been the subject of intense debate regarding its potential antiviral activity against SARS-Cov-2, the etiologic agent of COVID-19. Some report possible curative effects; others do not. Therefore, the objective of this study was to simulate possible scenarios of response to hydroxychloroquine in COVID-19 patients using mathematical modeling. METHODS: To shed some light on this controversial topic, we simulated hydroxychloroquine-based interventions on virus/host cell dynamics using a basic system of previously published differential equations. Mathematical modeling was implemented using Python programming language v 3.7. RESULTS: According to mathematical modeling, hydroxychloroquine may have an impact on the amplitude of the viral load peak and viral clearance if the drug is administered early enough (i.e., when the virus is still confined within the pharyngeal cavity). The effects of chloroquine/hydroxychloroquine may be fully explained only when also considering the capacity of this drug to increase the death rate of SARS-CoV-2-infected cells, in this case by enhancing the cell-mediated immune response. CONCLUSIONS: These considerations may not only be applied to chloroquine/hydroxychloroquine but may have more general implications for development of anti-COVID-19 combination therapies and prevention strategies through an increased death rate of the infected cells.


Subject(s)
Coronavirus Infections/drug therapy , Coronavirus Infections/prevention & control , Hydroxychloroquine/pharmacokinetics , Hydroxychloroquine/therapeutic use , Pandemics/prevention & control , Pneumonia, Viral/drug therapy , Pneumonia, Viral/prevention & control , Betacoronavirus/drug effects , COVID-19 , Chloroquine/pharmacokinetics , Chloroquine/therapeutic use , Coronavirus Infections/metabolism , Humans , Immunity, Cellular/drug effects , Models, Theoretical , Pneumonia, Viral/metabolism , SARS-CoV-2 , COVID-19 Drug Treatment
6.
Int J Antimicrob Agents ; 56(3): 106078, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32629115

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is having serious consequences on health and the economy worldwide. All evidence-based treatment strategies need to be considered to combat this new virus. Drugs need to be considered on scientific grounds of efficacy, safety and cost. Chloroquine (CQ) and hydroxychloroquine (HCQ) are old drugs used in the treatment of malaria. Moreover, their antiviral properties have been previously studied, including against coronaviruses, where evidence of efficacy has been found. In the current race against time triggered by the COVID-19 pandemic, the search for new antivirals is very important. However, consideration should be given to old drugs with known anti-coronavirus activity, such as CQ and HCQ. These could be integrated into current treatment strategies while novel treatments are awaited, also in light of the fact that they display an anticoagulant effect that facilitates the activity of low-molecular-weight heparin, aimed at preventing acute respiratory distress syndrome (ARDS)-associated thrombotic events. The safety of CQ and HCQ has been studied for over 50 years, however recently published data raise concerns for cardiac toxicity of CQ/HCQ in patients with COVID-19. This review also re-examines the real information provided by some of the published alarming reports, although concluding that cardiac toxicity should in any case be stringently monitored in patients receiving CQ/HCQ.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Chloroquine/therapeutic use , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Disseminated Intravascular Coagulation/prevention & control , Hydroxychloroquine/therapeutic use , Pneumonia, Viral/drug therapy , Angiotensin-Converting Enzyme 2 , Anti-Inflammatory Agents/therapeutic use , Anticoagulants/therapeutic use , Autophagy/drug effects , Autophagy/genetics , Betacoronavirus/growth & development , Betacoronavirus/immunology , COVID-19 , Coronavirus Infections/pathology , Coronavirus Infections/virology , Cytokine Release Syndrome/virology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Disseminated Intravascular Coagulation/virology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunologic Factors/therapeutic use , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2 , Virus Internalization/drug effects , Virus Replication/drug effects
7.
EMBO J ; 39(9): e102209, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32157726

ABSTRACT

HIV-1 persists in a latent form during antiretroviral therapy, mainly in CD4+ T cells, thus hampering efforts for a cure. HIV-1 infection is accompanied by metabolic alterations, such as oxidative stress, but the effect of cellular antioxidant responses on viral replication and latency is unknown. Here, we show that cells survive retroviral replication, both in vitro and in vivo in SIVmac-infected macaques, by upregulating antioxidant pathways and the intertwined iron import pathway. These changes are associated with remodeling of promyelocytic leukemia protein nuclear bodies (PML NBs), an important constituent of nuclear architecture and a marker of HIV-1 latency. We found that PML NBs are hyper-SUMOylated and that PML protein is degraded via the ubiquitin-proteasome pathway in productively infected cells, before latency establishment and after reactivation. Conversely, normal numbers of PML NBs were restored upon transition to latency or by decreasing oxidative stress or iron content. Our results highlight antioxidant and iron import pathways as determinants of HIV-1 latency and support their pharmacologic inhibition as tools to regulate PML stability and impair latency establishment.


Subject(s)
Gene Regulatory Networks , HIV Infections/virology , HIV-1/physiology , Iron/metabolism , Promyelocytic Leukemia Protein/metabolism , Animals , Cell Line , Disease Models, Animal , HIV Infections/genetics , HIV Infections/metabolism , Humans , Macaca , Oxidation-Reduction , Proteolysis , Sequence Analysis, RNA , Sumoylation , Up-Regulation , Virus Latency
8.
Commun Biol ; 3: 10, 2020.
Article in English | MEDLINE | ID: mdl-31909202

ABSTRACT

Gold compounds have a long history of use as immunosuppressants, but their precise mechanism of action is not completely understood. Using our recently developed liver-on-a-chip platform we now show that gold compounds containing planar N-heterocyclic carbene (NHC) ligands are potent ligands for the aryl hydrocarbon receptor (AHR). Further studies showed that the lead compound (MC3) activates TGFß1 signaling and suppresses CD4+ T-cell activation in vitro, in human and mouse T cells. Conversely, genetic knockdown or chemical inhibition of AHR activity or of TGFß1-SMAD-mediated signaling offsets the MC3-mediated immunosuppression. In scurfy mice, a mouse model of human immunodysregulation polyendocrinopathy enteropathy X-linked syndrome, MC3 treatment reduced autoimmune phenotypes and extended lifespan from 24 to 58 days. Our findings suggest that the immunosuppressive activity of gold compounds can be improved by introducing planar NHC ligands to activate the AHR-associated immunosuppressive pathway, thus expanding their potential clinical application for autoimmune diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Immunosuppression Therapy/methods , Organogold Compounds/immunology , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects , Transforming Growth Factor beta1/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/drug effects , Hep G2 Cells , Humans , Male , Mice , Receptors, Aryl Hydrocarbon/metabolism , Transforming Growth Factor beta1/metabolism
9.
Front Immunol ; 10: 2267, 2019.
Article in English | MEDLINE | ID: mdl-31636630

ABSTRACT

Recently, a second individual (the "London patient") with HIV-1 infection and concomitant leukemia was cured of both diseases by a conditioning myeloablative regimen followed by transplantation of stem cells bearing the homozygous CCR5 Δ32 mutation. The substantial risks and cost associated with this procedure render it unfeasible on a large scale. This strategy also indicates that a common pathway toward a cure for both HIV and cancer may exist. Successful approaches to curing both diseases should ideally possess three components, i.e., (1) direct targeting of pathological cells (neoplastic cells in cancer and the HIV-infected reservoir cells), (2) subsequent impediment to reconstitution of the pool of pathological cells and (3) sustained, immunologic control of the disease (both diseases are characterized by detrimental immune hyper-activation that hinders successful establishment of immunity). In this review, we explore medications that are either investigational or FDA-approved anticancer treatments that may be employed to achieve the goal of curing HIV-1. These include: thioredoxin reductase inhibitors (phases 1-3), immune checkpoint inhibitors (phases 1, 3), Jak inhibitors (FDA approved for arthritis and multiple cancer indications, summarized in Table 1). Of note, some of these medications such as arsenic trioxide and Jak inhibitors may also reversibly down regulate CCR5 expression on CD4+ T-cells, thus escaping the ethical issues of inducing or transferring mutations in CCR5 that are presently the subject of interest as it relates to HIV-1 cure strategies.


Subject(s)
HIV Infections/therapy , Neoplasms/therapy , Arsenic Trioxide/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , HIV Infections/immunology , Hematopoietic Stem Cell Transplantation , Humans , Janus Kinases/antagonists & inhibitors , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
10.
Int J Antimicrob Agents ; 54(5): 592-600, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31394172

ABSTRACT

Antiretroviral therapy (ART) is typically composed of a combination of three antiretroviral drugs and is the treatment of choice for people with human immunodeficiency virus type 1/acquired immune deficiency syndrome (HIV-1/AIDS). However, it is unable to impact on viral reservoirs, which harbour latent HIV-1 genomes that are able to reignite the infection upon treatment suspension. The aim of this study was to provide an estimate of the safety of the disease-modifying antirheumatic agent auranofin and its impact on the HIV-1 reservoir in humans under intensified ART. For this purpose, an interim analysis was conducted of three of the six arms of the NCT02961829 clinical trial (five patients each) with: no intervention, i.e. continuation of first-line ART; intensified ART (ART + dolutegravir and maraviroc); and intensified ART plus auranofin. Auranofin treatment was found to be well tolerated. No major adverse events were detected apart from a transient decrease in CD4+ T-cell counts at Weeks 8 and 12. Auranofin decreased total viral DNA in peripheral blood mononuclear cells compared with ART-only regimens at Week 20 (P = 0.036) and induced a decrease in integrated viral DNA as quantified by Alu PCR. Despite the limited number of patient-derived sequences available in this study, phylogenetic analyses of nef sequences support the idea that auranofin may impact on the viral reservoir. [ClinicalTrials.gov ID: NCT02961829].


Subject(s)
Antirheumatic Agents/therapeutic use , Auranofin/therapeutic use , HIV-1/genetics , Proviruses/drug effects , Proviruses/genetics , Virus Latency/drug effects , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , DNA, Viral/drug effects , DNA, Viral/genetics , HIV Fusion Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Maraviroc/therapeutic use , Oxazines , Piperazines , Pyridones
11.
Viruses ; 8(12)2016 11 29.
Article in English | MEDLINE | ID: mdl-27916837

ABSTRACT

Zika virus (ZIKV) infection in utero might lead to microcephaly and other congenital defects. Since no specific therapy is available thus far, there is an urgent need for the discovery of agents capable of inhibiting its viral replication and deleterious effects. Chloroquine is widely used as an antimalarial drug, anti-inflammatory agent, and it also shows antiviral activity against several viruses. Here we show that chloroquine exhibits antiviral activity against ZIKV in Vero cells, human brain microvascular endothelial cells, human neural stem cells, and mouse neurospheres. We demonstrate that chloroquine reduces the number of ZIKV-infected cells in vitro, and inhibits virus production and cell death promoted by ZIKV infection without cytotoxic effects. In addition, chloroquine treatment partially reveres morphological changes induced by ZIKV infection in mouse neurospheres.


Subject(s)
Antiviral Agents/pharmacology , Chloroquine/pharmacology , Endocytosis/drug effects , Zika Virus Infection/virology , Zika Virus/drug effects , Zika Virus/physiology , Animals , Cell Line , Chlorocebus aethiops , Humans , Mice
12.
J Clin Invest ; 126(5): 1630-9, 2016 05 02.
Article in English | MEDLINE | ID: mdl-27135880

ABSTRACT

Although the use of antioxidants for the treatment of cancer and HIV/AIDS has been proposed for decades, new insights gained from redox research have suggested a very different scenario. These new data show that the major cellular antioxidant systems, the thioredoxin (Trx) and glutathione (GSH) systems, actually promote cancer growth and HIV infection, while suppressing an effective immune response. Mechanistically, these systems control both the redox- and NO-based pathways (nitroso-redox homeostasis), which subserve innate and cellular immune defenses. Dual inhibition of the Trx and GSH systems synergistically kills neoplastic cells in vitro and in mice and decreases resistance to anticancer therapy. Similarly, the population of HIV reservoir cells that constitutes the major barrier to a cure for AIDS is exquisitely redox sensitive and could be selectively targeted by Trx and GSH inhibitors. Trx and GSH inhibition may lead to a reprogramming of the immune response, tilting the balance between the immune system and cancer or HIV in favor of the former, allowing elimination of diseased cells. Thus, therapies based on silencing of the Trx and GSH pathways represent a promising approach for the cure of both cancer and AIDS and warrant further investigation.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Glutathione/immunology , HIV-1/immunology , Neoplasms/immunology , Thioredoxins/immunology , Acquired Immunodeficiency Syndrome/therapy , Animals , Humans , Mice , Neoplasms/pathology , Neoplasms/therapy , Oxidation-Reduction
13.
Retrovirology ; 12: 51, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26084487

ABSTRACT

The restoration of the immune system prompted by antiretroviral therapy (ART) has allowed drastically reducing the mortality and morbidity of HIV infection. However, one main source of clinical concern is the persistence of immune hyperactivation in individuals under ART. Chronically enhanced levels of T-cell activation are associated with several deleterious effects which lead to faster disease progression and slower CD4(+) T-cell recovery during ART. In this article, we discuss the rationale, and review the results, of the use of antimalarial quinolines, such as chloroquine and its derivative hydroxychloroquine, to counteract immune activation in HIV infection. Despite the promising results of several pilot trials, the most recent clinical data indicate that antimalarial quinolines are unlikely to exert a marked beneficial effect on immune activation. Alternative approaches will likely be required to reproducibly decrease immune activation in the setting of HIV infection. If the quinoline-based strategies should nevertheless be pursued in future studies, particular care must be devoted to the dosage selection, in order to maximize the chances to obtain effective in vivo drug concentrations.


Subject(s)
Antimalarials/therapeutic use , Chloroquine/therapeutic use , Drug Repositioning , HIV Infections/immunology , HIV Infections/pathology , Hydroxychloroquine/therapeutic use , Immunologic Factors/therapeutic use , Humans , Treatment Outcome
14.
J Med Primatol ; 44(5): 227-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26058990

ABSTRACT

BACKGROUND: Administration of antiretroviral therapy and two experimental drugs, auranofin and buthionine sulfoximine (BSO), was previously shown to be followed by drug-free control of chronic SIVmac251 infection, decreased immune activation and increased cell-mediated anti-Gag responses. METHODS: Phylogeny was analysed with Phylogeny.fr. Entropy was calculated with the specific tool of the HIV Sequence Database. The capsid Gag structure was computed using SPDBV. The bottleneck effect was simulated through an appropriate online tool. RESULTS: The region of Gag predominantly targeted during control of SIVmac251 infection is highly conserved in primate lentiviruses and plays an important role in capsid architecture. Computer-aided simulations support the view that the preferential development of immune responses against this region is derived from a 'bottleneck effect' after restriction, by auranofin and BSO, of the activated lymphocyte pool. CONCLUSIONS: Restriction of immune activation through auranofin/BSO may result in stochastic selection of cell clones targeting conserved epitopes leading to a functional cure-like condition.


Subject(s)
Anti-Retroviral Agents/pharmacology , Auranofin/pharmacology , Buthionine Sulfoximine/pharmacology , Gene Products, gag/genetics , Immunity, Cellular/drug effects , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/genetics , Animals , Anti-Retroviral Agents/therapeutic use , Gene Products, gag/metabolism , Haplorhini , Molecular Sequence Data , Sequence Analysis, DNA , Simian Immunodeficiency Virus/drug effects
15.
J Virol ; 89(15): 7521-35, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972547

ABSTRACT

UNLABELLED: Off-therapy control of viremia by HIV-infected individuals has been associated with two likely players: a restricted viral reservoir and an efficient cell-mediated immune response. We previously showed that a combination of highly suppressive antiretroviral therapy and two experimental drugs, i.e., auranofin and buthionine sulfoximine, was able to reduce the viral reservoir, elicit efficient cell-mediated antiviral responses, and induce intermittent posttherapy viral load control in chronically SIVmac251-infected macaques. We here show that the macaques that had received this drug combination and then stopped antiretroviral therapy were also able to maintain low numbers of activated CD4+ T cells at viral rebound. Moreover, these macaques consistently displayed low-level simian immunodeficiency virus (SIV) diversity, which was in line with the strong and broadly reactive cell-mediated immune responses against conserved Gag antigens. Extended follow-up showed that the two macaques that had received the complete drug combination remained healthy and did not develop AIDS in 2 years of follow-up after therapy suspension. This disease-free survival is longer than twice the average time of progression to AIDS in SIVmac251-infected rhesus macaques. These results suggest that limited numbers of activated T cells at viral rebound and subsequent development of broadly reactive cell-mediated responses may be interrelated in reducing the viral reservoir. IMPORTANCE: The HIV reservoir in CD4+ T cells represents one main obstacle to HIV eradication. Recent studies, however, show that a drastic reduction of this reservoir is insufficient for inducing a functional cure of AIDS. In the present work, we thoroughly studied and subjected to long-term follow-up two macaques showing intermittent control of the virus following suspension of antiretroviral therapy plus an experimental antireservoir treatment, i.e., the gold salt auranofin and the investigational chemotherapeutic agent buthionione sulfoximine (BSO). We found that these drugs were able to decrease the number of activated CD4+ T cells, which are preferential targets for HIV infection. Then, efficient immune responses against the virus were developed in the macaques, which remained healthy during 2 years of follow-up. This result may furnish another building block for future attempts to cure HIV/AIDS.


Subject(s)
HIV Infections/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Animals , Antiviral Agents/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Disease Models, Animal , Follow-Up Studies , Gene Products, gag , HIV Infections/drug therapy , HIV Infections/virology , Humans , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Viral Load
16.
Retrovirology ; 10: 145, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24267982

ABSTRACT

Despite the huge clinical success of antiretroviral therapy, several factors such as side effects, requirement of life-long adherence, high cost, incomplete access to therapies and development of drug resistance make the quest for an ultimate cure of HIV/AIDS a worldwide priority of biomedical research. In this respect, several sterilizing or functional cures have been reported in the last years in both non-human primates and humans. This review provides a summary of the main results achieved so far, outlining their strengths as well as their limitations. A synthetic interpretation of these results could be pivotal in order to develop an effective and widely available cure.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , Anti-HIV Agents/therapeutic use , Biological Therapy/methods , Disease Eradication , Immunotherapy/methods , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans
17.
Retrovirology ; 10: 71, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23866829

ABSTRACT

BACKGROUND: HIV infection persists despite antiretroviral treatment (ART) and is reignited as soon as therapies are suspended. This vicious cycle is fueled by the persistence of viral reservoirs that are invulnerable to standard ART protocols, and thus therapeutic agents able to target these reservoirs are needed. One such agent, auranofin, has recently been shown to decrease the memory T-cell reservoir in chronically SIVmac251-infected macaques. Moreover, auranofin could synergize with a fully suppressive ART protocol and induce a drug-free post-therapy containment of viremia. RESULTS: We administered buthionine sulfoximine (BSO), an inhibitor of glutathione synthesis currently in clinical trials for cancer, in combination with auranofin to chronically SIVmac251-infected macaques under highly-intensified ART (H-iART). The ART/auranofin/BSO therapeutic protocol was followed, after therapy suspension, by a significant decrease of viral RNA and DNA in peripheral blood as compared to pre-therapy levels. Drug-free post-therapy control of the infection was achieved in animals with pre-therapy viral loads ranging from values comparable to average human set points to levels largely higher. This control was dependent on the presence CD8+ cells and associated with enhanced levels of cell-mediated immune responses. CONCLUSIONS: The level of post-therapy viral set point reduction achieved in this study is the largest reported so far in chronically SIVmac251-infected macaques and may represent a promising strategy to improve over the current "ART for life" plight.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Auranofin/administration & dosage , Immunity, Cellular , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Therapies, Investigational/methods , Withholding Treatment , Animals , Antiretroviral Therapy, Highly Active/methods , Buthionine Sulfoximine/administration & dosage , DNA, Viral/blood , Macaca , RNA, Viral/blood , Simian Immunodeficiency Virus/isolation & purification , Treatment Outcome , Viral Load
18.
Retrovirology ; 9: 75, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22978787

ABSTRACT

Our inability to cure HIV/AIDS is related to the ability of the virus to establish reservoirs during treatment. In order to develop new strategies, it is certainly essential that a suitable animal model be implemented. In the recent work of Shytaj et al., it has been possible to inhibit viral replication to levels below the assay detection limit in the macaque AIDS model. Moreover, different therapeutic regimens applied to the rhesus macaque AIDS model (herein reviewed), including ours, are starting to show the potential to induce, following therapy suspension, conditions reminiscent of a drug-free control of the infection.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , Anti-HIV Agents/administration & dosage , Animals , Disease Models, Animal , Humans , Macaca mulatta , Treatment Outcome
19.
PLoS Pathog ; 8(6): e1002774, 2012.
Article in English | MEDLINE | ID: mdl-22737073

ABSTRACT

Stably suppressed viremia during ART is essential for establishing reliable simian models for HIV/AIDS. We tested the efficacy of a multidrug ART (highly intensified ART) in a wide range of viremic conditions (10³-107) viral RNA copies/mL) in SIVmac251-infected rhesus macaques, and its impact on the viral reservoir. Eleven macaques in the pre-AIDS stage of the disease were treated with a multidrug combination (highly intensified ART) consisting of two nucleosidic/nucleotidic reverse transcriptase inhibitors (emtricitabine and tenofovir), an integrase inhibitor (raltegravir), a protease inhibitor (ritonavir-boosted darunavir) and the CCR5 blocker maraviroc. All animals stably displayed viral loads below the limit of detection of the assay (i.e. <40 RNA copies/mL) after starting highly intensified ART. By increasing the sensitivity of the assay to 3 RNA copies/mL, viral load was still below the limit of detection in all subjects tested. Importantly, viral DNA resulted below the assay detection limit (<2 copies of DNA/5*105 cells) in PBMCs and rectal biopsies of all animals at the end of the follow-up, and in lymph node biopsies from the majority of the study subjects. Moreover, highly intensified ART decreased central/transitional memory, effector memory and activated (HLA-DR⁺) effector memory CD4⁺ T-cells in vivo, in line with the role of these subsets as the main cell subpopulations harbouring the virus. Finally, treatment with highly intensified ART at viral load rebound following suspension of a previous anti-reservoir therapy eventually improved the spontaneous containment of viral load following suspension of the second therapeutic cycle, thus leading to a persistent suppression of viremia in the absence of ART. In conclusion, we show, for the first time, complete suppression of viral load by highly intensified ART and a likely associated restriction of the viral reservoir in the macaque AIDS model, making it a useful platform for testing potential cures for AIDS.


Subject(s)
Anti-HIV Agents/administration & dosage , Antiretroviral Therapy, Highly Active/methods , Simian Acquired Immunodeficiency Syndrome/drug therapy , T-Lymphocyte Subsets/drug effects , Viral Load/drug effects , Adenine/administration & dosage , Adenine/analogs & derivatives , Animals , Cyclohexanes/administration & dosage , Darunavir , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Therapy, Combination , Emtricitabine , Flow Cytometry , Fluorescent Antibody Technique , Macaca mulatta , Maraviroc , Organophosphonates/administration & dosage , Pyrrolidinones/administration & dosage , Raltegravir Potassium , Ritonavir/administration & dosage , Sulfonamides/administration & dosage , Tenofovir , Triazoles/administration & dosage , Viremia/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...