Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Comb Chem High Throughput Screen ; 24(5): 716-728, 2021.
Article in English | MEDLINE | ID: mdl-32798373

ABSTRACT

AIMS: To predict potential drugs for COVID-19 by using molecular docking for virtual screening of drugs approved for other clinical applications. BACKGROUND: SARS-CoV-2 is the betacoronavirus responsible for the COVID-19 pandemic. It was listed as a potential global health threat by the WHO due to high mortality, high basic reproduction number, and lack of clinically approved drugs and vaccines. The genome of the virus responsible for COVID-19 has been sequenced. In addition, the three-dimensional structure of the main protease has been determined experimentally. OBJECTIVE: To identify potential drugs that can be repurposed for treatment of COVID-19 by using molecular docking based virtual screening of all approved drugs. METHODS: A list of drugs approved for clinical use was obtained from the SuperDRUG2 database. The structure of the target in the apo form, as well as structures of several target-ligand complexes, were obtained from RCSB PDB. The structure of SARS-CoV-2 Mpro determined from X-ray diffraction data was used as the target. Data regarding drugs in clinical trials for COVID-19 was obtained from clinicaltrials.org. Input for molecular docking based virtual screening was prepared by using Obabel and customized python, bash, and awk scripts. Molecular docking calculations were carried out with Vina and SMINA, and the docked conformations were analyzed and visualized with PLIP, Pymol, and Rasmol. RESULTS: Among the drugs that are being tested in clinical trials for COVID-19, Danoprevir and Darunavir were predicted to have the highest binding affinity for the Main protease (Mpro) target of SARS-CoV-2. Saquinavir and Beclabuvir were identified as the best novel candidates for COVID-19 therapy by using Virtual Screening of drugs approved for other clinical indications. CONCLUSION: Protease inhibitors approved for treatment of other viral diseases have the potential to be repurposed for treatment of COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19/virology , Drug Evaluation, Preclinical , Molecular Docking Simulation , SARS-CoV-2/drug effects , Antiviral Agents/chemistry , Benzazepines/chemistry , Benzazepines/pharmacology , Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Darunavir/chemistry , Darunavir/pharmacology , Drug Repositioning , High-Throughput Screening Assays , Humans , Indoles/chemistry , Indoles/pharmacology , Isoindoles/chemistry , Isoindoles/pharmacology , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/pharmacology , Proline/analogs & derivatives , Proline/chemistry , Proline/pharmacology , Saquinavir/chemistry , Saquinavir/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology
2.
J Biochem Mol Toxicol ; 32(11): e22215, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30194790

ABSTRACT

Garlic has been used as a traditional medicine to treat various diseases. Garlic reduces the risk of some diseases. This protective effect is due to the organosulfur compounds of garlic. The aim of this study was to investigate the inhibition effects of garlic-derived compounds on human immunodeficiency virus type 1 (HIV-1) and as the most important anti-HIV-1 medicine. The activation of saquinavir is believed to be the principal mechanism behind the protective effects of HIV-1. Our theoretical calculations are performed for blood phase by using the density functional theory for the main compounds of garlic. The chemical activity and solubility of ajoene and the mainly derived compounds of garlic as theoretical calculations are important for the medical research comparing with the other compounds of the garlic. The theoretical calculations have helped us to determine which active ingredient of the garlic having inhibition effects on HIV-1 and saquinavir.


Subject(s)
Anti-HIV Agents/pharmacology , Garlic/chemistry , HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , HIV-1/drug effects , Phytochemicals/pharmacology , Saquinavir/antagonists & inhibitors , Anti-HIV Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Catalytic Domain , Computational Biology , Density Functional Theory , Disulfides/chemistry , Disulfides/pharmacology , Drug Interactions , Drug Synergism , HIV Protease/chemistry , HIV Protease Inhibitors/chemistry , HIV-1/enzymology , Molecular Docking Simulation , Phytochemicals/chemistry , Saquinavir/agonists , Saquinavir/chemistry , Saquinavir/pharmacology , Solubility , Sulfoxides
3.
J Cell Physiol ; 226(7): 1803-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506111

ABSTRACT

We have recently shown that covalent attachment of the NO moiety to the HIV protease inhibitor Saquinavir (Saq) produced a qualitatively new chemical entity, named Saquinavir-NO (Saq-NO), with enhanced anticancer properties and reduced toxicity. In this study we evaluated the impact of Saq-NO on the growth of A375 human melanoma cells, as a prototype of NO-dependent cancer model. The novel compound strongly affected the in vitro and in vivo progression of A375 melanoma cell growth. The mechanism of antimelanoma action comprised dual drug activity-induction of apoptotic cell death and acquisition of melanoma cell responsiveness to TRAIL. Saq-NO-triggered apoptosis was dependent on transient AKT up-regulation and reduced pERK and iNOS expression that were observed within the first 12 h of exposure to the drug. Thereafter, however, Saq-NO up-regulated both iNOS transcription and NO endogenous synthesis and sensitized A375 cells to TRAIL. Furthermore, reduced YY1 expression was observed after 24 h of Saq-NO exposure, which correlated with increased expression of DR5. The biological relevance of this complex and powerful action of Saq-NO was consistent with the marked drug-induced inhibition of the growth of A375 xenotransplants in nude mice.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Melanoma/drug therapy , Nitric Oxide Synthase Type II/metabolism , Saquinavir/analogs & derivatives , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Nude , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Saquinavir/pharmacology , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays , YY1 Transcription Factor/metabolism
4.
Antivir Ther ; 15(8): 1161-9, 2010.
Article in English | MEDLINE | ID: mdl-21149923

ABSTRACT

BACKGROUND: The effect of HIV type-1 (HIV-1) subtype on in vitro susceptibility and virological response to darunavir (DRV) and lopinavir (LPV) was studied using a broad panel of primary isolates, and in recombinant clinical isolates from treatment-naive, HIV-1-infected patients in the Phase III trial, AntiRetroviral Therapy with TMC114 ExaMined In naive Subjects (ARTEMIS). METHODS: Patients received DRV/ritonavir (DRV/r) 800/100 mg once daily (n=343) or LPV/ritonavir (LPV/r) 800/200 mg total daily dose (n=346), plus a fixed daily dose of emtricitabine and tenofovir disoproxil fumarate. RESULTS: DRV demonstrated high antiviral activity against a broad panel of HIV-1 major group (M) and outlier group (O) primary isolates in peripheral blood mononuclear cells, with a median 50% effective concentration (EC(50)) of 0.52 nM. Most (61%) patients in ARTEMIS harboured HIV-1 subtype B; other prevalent subtypes were C (13%) and CRF01_AE (17%); 9% harboured other subtypes. Median EC(50) values (interquartile range) for DRV were 1.79 nM (1.3-2.6) for subtype B, 1.12 nM (0.8-1.4) for C and 1.27 nM (1.0-1.7) for CRF01_AE. Virological response to DRV/r (HIV-1 RNA<50 copies/ml [intent-to-treat, time-to-loss of virological response algorithm]) was 81%, 87% and 85% for patients with subtype B, C and CRF01_AE infections, respectively. Similar results were observed in the LPV/r treatment group. CONCLUSIONS: In vitro susceptibility to DRV was comparable across HIV-1 subtypes in a broad panel of primary isolates and in recombinant clinical isolates. Once daily DRV/r 800/100 mg and LPV/r 800/200 mg were highly effective in ARTEMIS irrespective of the HIV-1 subtype studied, confirming their broad anti-HIV-1 activity.


Subject(s)
HIV Infections/virology , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Adamantane/pharmacology , Adult , Analysis of Variance , Atazanavir Sulfate , Carbamates/pharmacology , Darunavir , Drug Resistance, Viral , Furans , HIV Infections/drug therapy , HIV-1/classification , HIV-1/physiology , Humans , Indinavir/pharmacology , Lopinavir , Microbial Sensitivity Tests , Molecular Typing , Nelfinavir/pharmacology , Neuraminidase/antagonists & inhibitors , Oligopeptides/pharmacology , Pyridines/pharmacology , Pyrones/pharmacology , Saquinavir/pharmacology , Viral Load
5.
Biopharm Drug Dispos ; 31(8-9): 495-505, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21104925

ABSTRACT

BACKGROUND/AIMS: The aim of this study was to elucidate the impact of first-pass intestinal metabolism to therapeutic efficacy of antiretrovirals and to ascertain interaction mechanisms between garlic supplements (aged garlic extract) and HIV-protease inhibitors. METHODS: In vitro permeability through rat jejunum, Caco-2 cell monolayers was determined and CYP3A4 metabolism studies were performed. RESULTS: Saquinavir and darunavir efflux from enterocytes into gastrointestinal lumen significantly increased in the presence of aged garlic extract, whereas their CYP3A4 metabolism was inhibited. In the case of saquinavir a significant increase of its efflux was observed also in the presence of lower ritonavir doses. Because both drugs bound to different binding sites on P-glycoprotein and/or multidrug resistance associated protein 2 than garlic phytochemicals or ritonavir, lower amounts of antiretrovirals were absorbed. CONCLUSIONS: The fractions of tested anti-HIV drugs absorbed could decrease significantly during self-medication with garlic supplements or ritonavir dose adjustments. Due to distinct saquinavir and darunavir preferences for binding sites on efflux transporters, the presence of other compounds (garlic phytochemicals, ritonavir), capable of influencing intestinal transporter-enzyme interplay, might lead to pharmacokinetic interactions observed in clinical studies and case reports with anti-HIV drugs.


Subject(s)
Anti-HIV Agents/metabolism , Garlic , HIV Protease Inhibitors/metabolism , Intestinal Mucosa/metabolism , Plant Extracts/metabolism , Ritonavir/metabolism , Saquinavir/metabolism , Sulfonamides/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Biological Transport , Caco-2 Cells , Cytochrome P-450 CYP3A/metabolism , Darunavir , Dietary Supplements , Drug Interactions , Enterocytes/metabolism , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacology , Humans , Intestinal Absorption , Jejunum/metabolism , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Plant Extracts/pharmacokinetics , Plant Extracts/pharmacology , Rats , Rats, Wistar , Ritonavir/pharmacokinetics , Ritonavir/pharmacology , Saquinavir/pharmacokinetics , Saquinavir/pharmacology , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
6.
AAPS J ; 10(4): 577-86, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19082742

ABSTRACT

The objective was to directly compare the four different "calculation" methods of assessing P-gp inhibition potential using experimental data obtained from approximately 60 structurally diverse internal research and marketed compounds. Bidirectional studies for digoxin (probe for P-gp substrate) were performed with and without test compounds (at 10 microM). Four different calculation methods were applied to the same dataset (raw bidirectional permeability values) to obtain the "percent inhibition of P-gp" for these compounds using the different methods. Significantly different inhibition potential was obtained with the "exact" same experimental dataset depending on the calculation method used. Subsequently, entirely different conclusions regarding the "inhibition potential" of test compound was reached due to the different calculation methods. Based on the direct comparison of these methods, method no. 3 (i.e., inhibition of B to A permeability of digoxin) is recommended as the calculation method ideal during screening stages due to its high throughput amenability. The methodology is capable of rapidly screening compounds with adequate reliability for early stage drug discovery. Method no. 3 provides an abridged version of a bidirectional study that is fully capable of identifying all non-inhibitors (0-20%), moderate inhibitors (20-60%), and potent inhibitors (>60%) and demonstrates high correlation with method no. 1 (inhibition based on both A to B and B to A permeability of digoxin). Nevertheless, method no. 1 might be appropriate for more detailed mechanistic studies required in late stage discovery and development.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Caco-2 Cells/metabolism , Biological Transport/drug effects , Caco-2 Cells/drug effects , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Digoxin/pharmacokinetics , Digoxin/pharmacology , Drug Evaluation, Preclinical/methods , HIV Protease Inhibitors/pharmacology , Humans , Indinavir/pharmacology , Kinetics , Propionates/pharmacology , Quinolines/pharmacology , Saquinavir/pharmacology , Verapamil/pharmacology
7.
BMC Neurosci ; 9: 27, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18302767

ABSTRACT

BACKGROUND: Aspartyl protease inhibitors (PIs) used to treat HIV belong to an important group of drugs that influence significantly endothelial cell functioning and angiogenic capacity, although specific mechanisms are poorly understood. Recently, PIs, particularly Nelfinavir, were reported to disrupt Notch signaling in the HIV-related endothelial cell neoplasm, Kaposi's sarcoma. Given the importance of maintaining proper cerebral endothelial cell signaling at the blood brain barrier during HIV infection, we considered potential signaling pathways such as Notch, that may be vulnerable to dysregulation during exposure to PI-based anti-retroviral regimens. Notch processing by gamma-secretase results in cleavage of the notch intracellular domain that travels to the nucleus to regulate expression of genes such as vascular endothelial cell growth factor and NFkappaB that are critical in endothelial cell functioning. Since, the effects of HIV PIs on gamma-secretase substrate pathways in cerebral endothelial cell signaling have not been addressed, we sought to determine the effects of HIV PIs on Notch and amyloid precursor protein. RESULTS: Exposure to reported physiological levels of Saquinavir, Indinavir, Nelfinavir and Ritonavir, significantly increased reactive oxygen species in cerebral endothelial cells, but had no effect on cell survival. Likewise, PIs decreased Notch 4-protein expression, but had no effect on Notch 1 or amyloid precursor protein expression. On the other hand, only Nelfinavir increased significantly Notch 4 processing, Notch4 intracellular domain nuclear localization and the expression of notch intracellular domain targets NFkappaB and matrix metalloproteinase 2. Pre-treatment with the antioxidant Vitamin E prevented PI-induced reactive oxygen species generation and partially prevented Nelfinavir-induced changes in both Notch 4 processing, and cellular localization patterns. Moreover, in support of increased expression of pro-angiogenic genes after Nelfinavir treatment, Nelfinavir did not inhibit angiogenic capacity. CONCLUSION: Nelfinavir affects Notch 4 processing that results in induction of expression of the pro-angiogenic genes NFkappaB and matrix metalloproteinase 2 in cerebral endothelial cells.


Subject(s)
Endothelial Cells/drug effects , HIV Protease Inhibitors/pharmacology , Nelfinavir/pharmacology , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Receptors, Notch/drug effects , Receptors, Notch/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Cerebral Arteries/cytology , Cerebral Veins/cytology , Frontal Lobe/blood supply , Frontal Lobe/cytology , Humans , Indinavir/pharmacology , Oxidative Stress/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Receptor, Notch1/metabolism , Receptor, Notch4 , Repressor Proteins/metabolism , Ritonavir/pharmacology , Saquinavir/pharmacology , Vitamin E/pharmacology
8.
Can J Physiol Pharmacol ; 85(9): 966-78, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18066144

ABSTRACT

In this study, we used an in vitro Caco-2 cell monolayer model to evaluate aqueous extracts of commercial-source goldenseal (Hydrastis canadensis) and milk thistle (Silybum marianum) capsule formulations, their marker phytochemicals (berberine and silibinin, respectively), as well as dillapiol, vinblastine, and the HIV protease inhibitor saquinavir for their ability to modulate CYP3A4 and ABCB1 expression after short-term exposure (48 h). Both upregulation and downregulation of CYP3A4 expression was observed with extracts of varying concentrations of the two natural health products (NHPs). CYP3A4 was highly responsive in our system, showing a strong dose-dependent modulation by the CYP3A4 inhibitor dillapiol (upregulation) and the milk thistle flavonolignan silibinin (downregulation). ABCB1 was largely unresponsive in this cellular model and appears to be of little value as a biomarker under our experimental conditions. Therefore, the modulation of CYP3A4 gene expression can serve as an important marker for the in vitro assessment of NHP-drug interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Hydrastis , Silybum marianum , ATP Binding Cassette Transporter, Subfamily B , Allyl Compounds , Antineoplastic Agents, Phytogenic/pharmacology , Caco-2 Cells , Capsules , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Dioxoles/pharmacology , HIV Protease Inhibitors/pharmacology , Humans , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Saquinavir/pharmacology , Vinblastine/pharmacology
9.
Microbes Infect ; 8(7): 1783-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16815068

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) protease (PR) plays an essential role in processing viral polyproteins into mature proteins. As a result, it is a major target for the development of drugs against AIDS. However, due to the rapid emergence of drug-resistant HIV, the development of novel HIV PR inhibitors is urgently needed. We recently established a new cell line E-PR293 which can be used as a safe, convenient and highly efficient assay system to screen HIV-1 PR inhibitors. In the cells, the HIV-1 PR is expressed in a chimeric protein with the green fluorescence protein (GFP). This assay measures the PR activity as a function of either the fluorescence of GFP or the cytotoxic activity of HIV-1 PR which is expressed in the cell. E-PR293 cells were maintained in the presence of doxycycline, which suppresses the expression of HIV-1 PR. The removal of doxycycline induces the expression of HIV-1 PR, which is used to screen HIV-1 PR inhibitors. In E-PR293 cells, the 50% inhibitory concentration of the cytotoxic effects by nelfinavir and saquinavir were as low as nanomolar levels, almost equal to those found in the HIV-infection assay.


Subject(s)
Cell Line , Drug Evaluation, Preclinical/methods , HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , Doxycycline/pharmacology , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HIV Protease/biosynthesis , HIV Protease/genetics , Humans , Nelfinavir/pharmacology , Recombinant Fusion Proteins/biosynthesis , Saquinavir/pharmacology , Sensitivity and Specificity
10.
HIV Clin Trials ; 2(6): 445-52, 2001.
Article in English | MEDLINE | ID: mdl-11742431

ABSTRACT

PURPOSE: To relate baseline plasma HIV genotypic and virtual phenotypic antiretroviral drug susceptibility to subsequent virological response in patients receiving saquinavir (SQV)-enhancing therapy. Individuals were randomized to receive stavudine (d4T), SQV, and one of ritonavir, nelfinavir, or delavirdine to enhance SQV blood levels. METHOD: The protease and reverse transcriptase baseline sequences of 31 treatment-experienced patients were analyzed by genotype and virtual phenotype and were related to viral load at weeks 12 and 24. Genotypic resistance to SQV was defined by the presence of G48V and/or L90M mutations in the protease gene. Potential cross-resistance to d4T in zidovudine (ZDV)-experienced individuals was defined by the presence of thymidine-associated mutations in the reverse transcriptase gene. RESULTS: ZDV-associated mutations did not affect the virological response at 24 weeks. Individuals who were sensitive to SQV at baseline as determined by either genotyping or virtual phenotyping showed a greater decrease in viral load at week 24 than those resistant to SQV, irrespective of treatment arm. By genotyping, SQV-sensitive individuals had a median log decrease of 1.12 compared to 0.32 for those individuals who were SQV resistant. By virtual phenotyping, SQV-sensitive individuals had a median log decrease of 1.0 compared to a rise of 0.08 in resistant individuals. CONCLUSION: Thymidine analogue-associated mutations at baseline did not influence the response to subsequent therapy involving d4T. Individuals who were sensitive or resistant to SQV by genotyping or virtual phenotyping responded to SQV-enhancing regimens, but the virological response was greater in those who were sensitive.


Subject(s)
Anti-HIV Agents/therapeutic use , Drug Resistance, Viral , HIV Infections/drug therapy , HIV-1/drug effects , Reverse Transcriptase Inhibitors/therapeutic use , Saquinavir/therapeutic use , Anti-HIV Agents/pharmacology , Drug Therapy, Combination , Genotype , HIV Infections/virology , HIV Protease/genetics , HIV Reverse Transcriptase/genetics , HIV-1/genetics , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Phenotype , RNA, Viral/blood , Reverse Transcriptase Inhibitors/pharmacology , Saquinavir/pharmacology , Treatment Outcome , Viral Load
11.
Blood ; 96(10): 3553-9, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071654

ABSTRACT

Inhibitors of the protease of human immunodeficiency virus type 1 (HIV-1) may inhibit cytoplasmic retinoic acid-binding proteins, cytochrome P450 isoforms, as well as P-glycoproteins. These features of the protease inhibitors might enhance the activity of retinoids. To explore this hypothesis, myeloid leukemia cells were cultured with all-trans retinoic acid (ATRA) either alone or in combination with the HIV-1 protease inhibitors indinavir, ritonavir, and saquinavir. Consistent with the hypothesis, the HIV-1 protease inhibitors enhanced the ability of ATRA to inhibit growth and induce differentiation of HL-60 and NB4 myeloid leukemia cells, as measured by expression of CD11b and CD66b cell surface antigens, as well as reduction of nitroblue tetrazolium. Growth of ATRA-resistant UF-1 cells was also inhibited when cultured with the combination of ATRA and indinavir. Moreover, indinavir enhanced the ability of ATRA to induce expression of the myeloid differentiation-related transcription factor C/EBPepsilon messenger RNA in NB4 cells by 9.5-fold. Taken together, the results show that HIV-1 protease inhibitors enhance the antiproliferative and differentiating effects of ATRA on myeloid leukemia cells. An HIV-1 protease inhibitor might be a useful adjuvant with ATRA for patients with acute promyelocytic leukemia and possibly retinoid-resistant cancers.


Subject(s)
Cell Differentiation/drug effects , HIV Protease Inhibitors/pharmacology , Leukemia, Myeloid/drug therapy , CCAAT-Enhancer-Binding Proteins/genetics , Cell Division/drug effects , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Immunophenotyping , Indinavir/pharmacology , Leukemia, Myeloid/pathology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/pathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Ritonavir/pharmacology , Saquinavir/pharmacology , Tretinoin/pharmacology , Tumor Cells, Cultured
12.
Antiviral Res ; 42(3): 177-87, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10443530

ABSTRACT

HIV-host infection systems in vitro are important in the pre-clinical assessment of anti-retroviral drug activity. The present report describes the development of a new HIV-host model comprised of an epithelial cell line of HeLa lineage (HeLa-1), transfected with expression vectors bearing tat and rev (TART) genes of HIV-1 as well as the CD4 receptor gene, and HIV-1(delta Tat/Rev), a biologically contained strain of HIV-1 deleted in tat and rev. Measurement of infectivity, by syncytium formation and reverse transcriptase assay, revealed that HeLa-1 is infected with HIV-1(deltaTat/Rev). This virus failed to productively infect the TART-deficient CD4-positive HeLa cells, confirming its contained, non-infectious nature. The HeLa-1/HIV-1deltaTat/Rev system was used to measure the anti-retroviral activity of a human leukocyte-derived interferon (IFN-alphan3) preparation, several nucleoside analogs, and protease inhibitors. The HeLa-1/ HIV-1(deltaTat/Rev model provides a biologically contained system for the study of the HIV pathogenesis and the relative and combined therapeutic effects of anti-retroviral agents in vitro.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Evaluation, Preclinical/methods , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Interferon-alpha/pharmacology , Nelfinavir/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , CD4 Antigens/genetics , Gene Expression , Gene Products, rev/genetics , Gene Products, tat/genetics , HeLa Cells , Humans , In Vitro Techniques , Ritonavir/pharmacology , Saquinavir/pharmacology , Zidovudine/pharmacology , rev Gene Products, Human Immunodeficiency Virus , tat Gene Products, Human Immunodeficiency Virus
13.
J Med Virol ; 56(4): 332-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9829638

ABSTRACT

During a randomized double-blind study to assess the antiviral activity of saquinavir (SQV) alone or in combination with zidovudine (ZDV), the emergence of phenotypic resistance was evaluated in 44 patients treated with SQV (13 subjects), ZDV (14 subjects), and SQV plus ZDV (17 subjects). A significant (P< 0.05) lower CD4+ cell count and higher HIV RNA copy number at entry were found in six patients who developed resistant viral strain (3 to ZDV and 3 to SQV) during the first 4 months of treatment. After 1 year, drug-resistant strains (12 to ZDV and 14 to SQV) were isolated in 26 out of 37 patients. A significant higher number of patients treated with ZDV alone (10/13) harbored ZDV-resistant strains compared to patients treated by combination therapy (2/13); whereas more than 50% of patients had SQV-resistant strains aside from treatment. Early SQV-resistant strains were isolated in a limited number of patients treated with SQV alone (3/13). The rates of emergence of resistant strains during ZDV or SQV monotherapies are comparable. Combination therapy may delay the emergence of phenotypic resistance to either drugs in the short term and to ZDV, but not to SQV, at least after 1 year.


Subject(s)
HIV Infections/drug therapy , HIV-1/drug effects , Saquinavir/pharmacology , Zidovudine/pharmacology , Anti-HIV Agents/pharmacology , CD4 Lymphocyte Count , Double-Blind Method , Drug Resistance, Microbial , Drug Therapy, Combination , HIV Infections/virology , HIV-1/isolation & purification , Humans , Microbial Sensitivity Tests , RNA, Viral/blood , Saquinavir/therapeutic use , Zidovudine/therapeutic use
14.
J Med Chem ; 41(18): 3387-401, 1998 Aug 27.
Article in English | MEDLINE | ID: mdl-9719591

ABSTRACT

On the basis of previously described X-ray studies of an enzyme/aza-dipeptide complex,8 aza-dipeptide analogues carrying N-(bis-aryl-methyl) substituents on the (hydroxethyl)hydrazine moiety have been designed and synthesized as HIV-1 protease inhibitors. By using either equally (12) or orthogonally (13) protected dipeptide isosteres, symmetrically and asymmetrically acylated aza-dipeptides can be synthesized. This approach led to the discovery of very potent inhibitors with antiviral activities (ED50) in the subnanomolar range. Acylation of the (hydroxethyl)hydrazine dipeptide isostere with the L-tert-leucine derivative 29 increased the oral bioavailability significantly when compared to the corresponding L-valine or L-isoleucine derivatives. The bis(L-tert-leucine) derivatives CGP 75355, CGP 73547, CGP 75136, and CGP 75176 combine excellent antiviral activity with high blood concentration after oral administration. Furthermore, they show no cross-resistance with saquinavir-resistant strains and maintain activity against indinavir-resistant ones. Consequently they qualify for further profiling as potential clinical candidates.


Subject(s)
Anti-HIV Agents , Aza Compounds , Dipeptides , HIV Protease Inhibitors , HIV Protease/metabolism , Administration, Oral , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Aza Compounds/administration & dosage , Aza Compounds/chemical synthesis , Aza Compounds/pharmacology , Biological Availability , Dipeptides/administration & dosage , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Microbial , Female , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/enzymology , HIV-1/physiology , Indinavir/pharmacology , Mice , Mice, Inbred BALB C , Saquinavir/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL