Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
AIDS Res Hum Retroviruses ; 39(9): 475-481, 2023 09.
Article in English | MEDLINE | ID: mdl-37053110

ABSTRACT

Non-small cell lung cancer (NSCLC) is the most fatal non-AIDS defining cancer in people living with HIV (PWH) on antiretroviral therapy (ART). Treatment of malignancies in PWH requires concomitant cancer therapy and ART, which can lead to potential drug-drug interactions (DDIs) and overlapping toxicities. In this study, we hypothesize that replacement of ART with HIV broadly neutralizing antibodies (bNAbs) during cancer chemotherapy (chemo) may maintain HIV suppression and tumor inhibition while minimizing DDIs and overlapping toxicities. We compared HIV suppression, tumor inhibition, and toxicity between conventional treatment (ART plus chemo) and a new modality (bNAbs plus chemo) in humanized mice. Humanized mice infected with HIVYU2 and xenografted with human NSCLC A549 cells were treated with NSCLC chemo (cisplatin and gemcitabine) and first-line ART (dolutegravir, tenofovir disoproxil difumarate, and emtricitabine) or bNAbs (N49P9.6-FR and PGT 121) at human equivalent drug doses. We monitored plasma HIV RNA, tumor volume, and toxicities over five cycles of chemo. We found that chemo plus ART or bNAbs were equally effective at maintaining suppression of HIV viremia and tumor growth. Comparative analysis showed that mice on ART and chemo had significant reductions in body weight and significant increases in plasma creatinine concentrations compared with mice on bNAbs and chemo, which suggests that a combination of bNAbs and chemo produces less renal toxicity than an ART and chemo combination. These data suggest that bNAb therapy during concomitant chemo may be an improved treatment option over ART for PWH and NSCLC, and possibly other cancers, because bNAbs maintain HIV suppression while minimizing DDIs and toxicities.


Subject(s)
Carcinoma, Non-Small-Cell Lung , HIV Infections , HIV-1 , Lung Neoplasms , Humans , Mice , Animals , HIV Infections/drug therapy , Broadly Neutralizing Antibodies/pharmacology , Broadly Neutralizing Antibodies/therapeutic use , HIV Antibodies , Carcinoma, Non-Small-Cell Lung/drug therapy , Antibodies, Neutralizing , Lung Neoplasms/drug therapy , HIV-1/genetics
2.
Methods Mol Biol ; 2407: 229-251, 2022.
Article in English | MEDLINE | ID: mdl-34985669

ABSTRACT

Combination antiretroviral therapy (cART) suppresses HIV in most patients, but it cannot cure HIV infection. The main challenge to a cure is the presence of latent replication-competent HIV in resting CD4+ T cells in blood and tissues, which reignite infection after cART removal. The long half-life of this reservoir is a major barrier to a cure, and its elimination is a main goal of current HIV research. Animal models that recapitulate HIV latency can provide key insights into the establishment of HIV latency and, more importantly, enable the testing of HIV eradication strategies. We describe a protocol for the generation of humanized mice by intrahepatic injection of human cord blood-derived CD34+ hematopoietic stem cells (HSC) into newborn NSG mice, the HSC-NSG mouse model. We also describe a protocol for establishing HIV latency in this model. HSC-NSG mice have provided proof-of-concept for an approach combining HIV gene editing and HIV suppression in tissues that may cure HIV in infected humans.


Subject(s)
HIV Infections , HIV-1 , Animals , CD4-Positive T-Lymphocytes , HIV-1/genetics , Hematopoietic Stem Cells , Humans , Mice , Virus Latency
4.
Antivir Ther ; 24(3): 177-186, 2019.
Article in English | MEDLINE | ID: mdl-30574873

ABSTRACT

BACKGROUND: Cancer is a leading cause of death in HIV-infected patients in the era of combination antiretroviral therapy (cART). Yet, there are no specific guidelines for the combined use of cART and chemotherapy in HIV-infected cancer patients. The cellular enzyme thymidylate synthase (TS) catalyses the conversion of dUMP to TMP, which is converted to TDP and ultimately to TTP, a building block in DNA synthesis. TS inhibitors are recommended in some cancers, particularly non-small cell lung cancer (NSCLC). Because TS inhibitors modulate intracellular concentrations of endogenous 2'-deoxynucleotides, we hypothesized that TS inhibitors could impact the anti-HIV activity of nucleoside analogue reverse transcriptase inhibitors (NRTIs). METHODS: We evaluated gemcitabine and pemetrexed, two approved TS inhibitors, on the anti-HIV activities of NRTIs in infectivity assays using peripheral blood mononuclear cells (PBMCs) and in humanized mice. RESULTS: Gemcitabine enhanced the anti-HIV activities of tenofovir, abacavir and emtricitabine (FTC) in PBMCs. In contrast, pemetrexed had no effect on tenofovir, enhanced abacavir and, unexpectedly, decreased FTC and lamivudine (3TC) activities. Pemetrexed inhibitory effects on FTC and 3TC may be due to lower concentrations of active metabolites (FTCtp and 3TCtp) relative to their competing endogenous nucleotide (dCTP), as shown by decreases in FTCtp/dCTP ratios. Gemcitabine enhanced tenofovir while pemetrexed abrogated FTC antiviral activity in humanized mice. CONCLUSIONS: Chemotherapy with TS inhibitors can have opposing effects on cART, potentially impacting control of HIV and thereby development of viral resistance and size of the reservoir in HIV-infected cancer patients. Combinations of cART and chemotherapy should be carefully selected.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antiretroviral Therapy, Highly Active , Drug Interactions , HIV Infections/complications , HIV Infections/drug therapy , Neoplasms/complications , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antiretroviral Therapy, Highly Active/methods , Biomarkers , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , HIV Infections/immunology , HIV Infections/virology , Humans , Male , Mice , Mice, Transgenic , Treatment Outcome , Viral Load
5.
PLoS One ; 12(8): e0183425, 2017.
Article in English | MEDLINE | ID: mdl-28817720

ABSTRACT

Successful propagation of HIV in the human host requires entry into a permissive cell, reverse transcription of viral RNA, integration into the human genome, transcription of the integrated provirus, and assembly/release of new virus particles. Currently, there are antiretrovirals against each of these viral steps, except for provirus transcription. An inhibitor of HIV transcription could both increase potency of treatment and suppress drug-resistant strains. Cellular cyclin-dependent kinase 9 (CDK9) serves as a cofactor for the HIV Tat protein and is required for effective transcription of the provirus. Previous studies have shown that the CDK9 inhibitor Indirubin 3'-monoxime (IM) inhibits HIV transcription in vitro and in short-term in vivo studies of HIV acute infection in humanized mice (PBMC-NSG model), suggesting a therapeutic potential. The objective of this study is to evaluate the toxicity, pharmacokinetics and long-term antiviral activity of IM during chronic HIV infection in humanized mice (HSC-NSG model). We show that IM concentrations above EC50 values are rapidly achieved and sustained for > 3 h in plasma, and that non-toxic concentrations durably reduce HIV RNA levels. In addition, IM enhanced the antiviral activity of antiretrovirals from the reverse transcriptase, protease and integrase inhibitor classes in in vitro infectivity assays. In summary, IM may enhance current antiretroviral treatments and could help achieve a "functional cure" in HIV patients by preventing expression of proviruses.


Subject(s)
Anti-HIV Agents/pharmacology , Cyclin-Dependent Kinase 9/antagonists & inhibitors , HIV-1/isolation & purification , Indoles/pharmacology , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , Viral Load/drug effects , Viremia/prevention & control , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/toxicity , Cyclin-Dependent Kinase 9/metabolism , Humans , Indoles/pharmacokinetics , Indoles/toxicity , Mice , Oximes/pharmacokinetics , Oximes/toxicity
6.
J Antimicrob Chemother ; 72(9): 2570-2573, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28637235

ABSTRACT

Objectives: To compare the effectiveness of HIV integrase inhibitor monotherapy between raltegravir and dolutegravir as an approach to simplify therapy. Methods: We evaluated and compared the efficacy of 20 week monotherapy with dolutegravir or raltegravir in humanized mice (HSC-NSG) infected with HIVBaL. Plasma HIV RNA was measured by quantitative RT-PCR (limit of detection of 150 copies/45 µL of plasma) and drug levels by LC-MS/MS. Escape viruses were genotyped and analysed for replication capacity and drug susceptibility in tissue culture. Results: Drug-untreated control mice maintained constant viraemia throughout the study. Virus isolates from these mice were susceptible to both raltegravir (EC50 of <8 nM) and dolutegravir (EC50 of <1 nM). Mice treated with raltegravir or dolutegravir had plasma drug levels comparable to those in humans. Monotherapy with raltegravir initially suppressed HIV viraemia, but failed to maintain suppression in 4/4 mice. Viruses from raltegravir failing mice developed mutations G140G/S and Q148H/K, and were resistant to both raltegravir (EC50 values of >100 nM) and dolutegravir (EC50 values ranging from 8.8 to 13.3 nM). Monotherapy with dolutegravir suppressed viraemia in 5/5 of mice, but viraemia rebounded in one animal. The virus from this mouse had mutations E138K, G140S, Q148H, N155H and S230R, was highly resistant to both raltegravir (EC50 of >1000 nM) and dolutegravir (EC50 of 550 nM), and replicated to levels similar to those of control viruses in PBMCs. Conclusions: Monotherapy with either raltegravir or dolutegravir does not consistently maintain HIV suppression, suggesting that dual therapy may be required in simplification strategies.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/therapeutic use , Raltegravir Potassium/therapeutic use , Viremia/drug therapy , Animals , Genotype , HIV Infections/virology , HIV Integrase Inhibitors/therapeutic use , HIV-1/genetics , Humans , Mice , Mice, Transgenic , Mutation , Oxazines , Piperazines , Pyridones , RNA, Viral/blood , Viremia/virology , Virus Replication/drug effects
8.
AIDS ; 28(3): 317-23, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24326355

ABSTRACT

OBJECTIVE: The M184V mutation in the HIV-1 reverse transcriptase gene is frequent (>50%) in patients, both in resource-rich and resource-limited countries, conferring high-level resistance (>100-fold) to the cytosine analog reverse transcriptase inhibitors lamivudine and emtricitabine. The reverse transcriptase enzyme of M184V HIV-1 mutants has reduced processivity, resulting in reduced viral replication, particularly at low deoxynucleotide (dNTP) levels. We hypothesized that lowering intracellular dNTPs with resveratrol, a dietary supplement, could interfere with replication of M184V HIV-1 mutants. DESIGN AND METHODS: Evaluation of the activity of resveratrol on infection of primary peripheral blood lymphocytes by wild-type and M184V mutant HIV-1. We assayed both molecular clones and primary isolates of HIV-1, containing M184V alone and in combination with other reverse transcriptase mutations. Viral infection was quantified by p24 ELISA and by quantitative real-time PCR analysis. Cell viability was measured by colorimetric 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assays. RESULTS: In virus-infectivity assays, resveratrol did not inhibit replication of wild-type NL4-3 (resveratrol EC50 > 10 µmol/l), but it inhibited NL4-3 184V mutant (resveratrol EC50 = 5.8 µmol/l). These results were confirmed by real-time PCR analysis of early and late products of reverse transcription. Resveratrol inhibited molecular clones and primary isolates carrying M184V, alone or in combination with other reverse transcriptase mutations (resveratrol EC50 values ranging from 2.5 to 7.7 µmol/l). CONCLUSIONS: Resveratrol inhibits HIV-1 strains carrying the M184V mutation in reverse transcriptase. We propose resveratrol as a potential adjuvant in HIV-1 therapy, particularly in resource-limited settings, to help control emtricitabine-resistant M184V HIV-1 mutants.


Subject(s)
Anti-HIV Agents/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Viral , HIV-1/drug effects , HIV-1/physiology , Stilbenes/pharmacology , Virus Replication/drug effects , Cells, Cultured , Deoxycytidine/pharmacology , Emtricitabine , Enzyme-Linked Immunosorbent Assay , HIV Core Protein p24/analysis , HIV Reverse Transcriptase/genetics , HIV-1/growth & development , Humans , Lymphocytes/drug effects , Lymphocytes/virology , Mutation, Missense , Real-Time Polymerase Chain Reaction , Resveratrol
9.
J Infect Dis ; 208(12): 2085-94, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23922365

ABSTRACT

BACKGROUND: Targeting host-cell pathways to increase the potency of nucleoside/nucleotide analog reverse transcriptase inhibitors (NRTIs) is an important strategy for clinical investigation. Resveratrol is a natural product that inhibits cellular ribonucleotide reductase, prolonging the S phase of the cell cycle and preferentially lowering dATP levels. METHODS: We performed in vitro evaluation of resveratrol on the antiviral activity of adenosine analog tenofovir (TFV) against sensitive and drug-resistant human immunodeficiency virus type 1 (HIV-1), from subtypes B and C, in primary cells. RESULTS: Resveratrol enhanced the antiviral activity of TFV by up to 10-fold and restored susceptibility of TFV-resistant viruses. Resveratrol prevented wild-type HIV-1 from developing phenotypic resistance to TFV. Notably, resveratrol enhanced TFV activity against sensitive and resistant HIV-1 from both subtypes B and C. CONCLUSIONS: Prolonged wide-scale use of thymidine analogs in the setting of viral failure has limited the efficacy of second-line NRTI-based regimens in Africa. Moreover, the extensive use of ddI and d4T has led to high frequencies of the K65R mutation, further compromising TFV efficacy. In light of increasing resistance to commonly used NRTIs in global HIV treatment programs, targeting nucleoside biosynthesis with resveratrol, or derivatives with improved bioavailabilities, is a potential strategy to maintain, enhance, and restore susceptibility of commonly used NRTIs.


Subject(s)
Adenine/analogs & derivatives , HIV-1/metabolism , Organophosphonates/pharmacology , Purines/biosynthesis , Reverse Transcriptase Inhibitors/pharmacology , Stilbenes/pharmacology , Adenine/pharmacology , Cells, Cultured , Drug Resistance, Viral , Drug Synergism , HIV-1/drug effects , Host-Pathogen Interactions/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Purines/metabolism , Receptors, CCR5 , Resveratrol , Tenofovir , Virus Replication/drug effects
10.
Virology ; 411(1): 32-40, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21232779

ABSTRACT

R5 HIV-1 strains resistant to the CCR5 antagonist Maraviroc (MVC) can use drug-bound CCR5. We demonstrate that MVC-resistant HIV-1 exhibits delayed kinetics of coreceptor engagement and fusion during drug-bound versus free CCR5 infection of cell lines. Antibodies directed against the second extracellular loop (ECL2) of CCR5 had greater antiviral activity against MVC-bound compared to MVC-free CCR5 infection. However, in PBMCs, only ECL2 CCR5 antibodies HGS004 and HGS101, but not 2D7, inhibited infection by MVC resistant HIV-1 more potently with MVC-bound than with free CCR5. In addition, HGS004 and HGS101, but not 2D7, restored the antiviral activity of MVC against resistant virus in PBMCs. In flow cytometric studies, CCR5 binding by the HGS mAbs, but not by 2D7, was increased when PBMCs were treated with MVC, suggesting MVC increases exposure of the relevant epitope. Thus, HGS004 and HGS101 have antiviral mechanisms distinct from 2D7 and could help overcome MVC resistance.


Subject(s)
Anti-HIV Agents/pharmacology , Antibodies/immunology , Cyclohexanes/pharmacology , Drug Resistance, Viral , HIV-1/physiology , Receptors, CCR5/immunology , Triazoles/pharmacology , Virus Internalization , CCR5 Receptor Antagonists , Cells, Cultured , HIV-1/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Maraviroc , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...