Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Int J Mol Sci ; 23(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36293006

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) has continued to be a global concern. With the new HIV incidence, the emergence of multi-drug resistance and the untoward side effects of currently used anti-HIV drugs, there is an urgent need to discover more efficient anti-HIV drugs. Modern computational tools have played vital roles in facilitating the drug discovery process. This research focuses on a pharmacophore-based similarity search to screen 111,566,735 unique compounds in the PubChem database to discover novel HIV-1 protease inhibitors (PIs). We used an in silico approach involving a 3D-similarity search, physicochemical and ADMET evaluations, HIV protease-inhibitor prediction (IC50/percent inhibition), rigid receptor-molecular docking studies, binding free energy calculations and molecular dynamics (MD) simulations. The 10 FDA-approved HIV PIs (saquinavir, lopinavir, ritonavir, amprenavir, fosamprenavir, atazanavir, nelfinavir, darunavir, tipranavir and indinavir) were used as reference. The in silico analysis revealed that fourteen out of the twenty-eight selected optimized hit molecules were within the acceptable range of all the parameters investigated. The hit molecules demonstrated significant binding affinity to the HIV protease (PR) when compared to the reference drugs. The important amino acid residues involved in hydrogen bonding and п-п stacked interactions include ASP25, GLY27, ASP29, ASP30 and ILE50. These interactions help to stabilize the optimized hit molecules in the active binding site of the HIV-1 PR (PDB ID: 2Q5K). HPS/002 and HPS/004 have been found to be most promising in terms of IC50/percent inhibition (90.15%) of HIV-1 PR, in addition to their drug metabolism and safety profile. These hit candidates should be investigated further as possible HIV-1 PIs with improved efficacy and low toxicity through in vitro experiments and clinical trial investigations.


Subject(s)
Anti-HIV Agents , HIV Protease Inhibitors , HIV-1 , Humans , HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , Darunavir/pharmacology , Indinavir/chemistry , Indinavir/metabolism , Indinavir/pharmacology , Nelfinavir/chemistry , Nelfinavir/metabolism , Nelfinavir/pharmacology , Ritonavir/chemistry , Saquinavir/metabolism , Saquinavir/pharmacology , Lopinavir/pharmacology , Atazanavir Sulfate/pharmacology , Molecular Docking Simulation , Anti-HIV Agents/pharmacology , Amino Acids/pharmacology
2.
Mol Pharmacol ; 99(3): 184-196, 2021 03.
Article in English | MEDLINE | ID: mdl-33483427

ABSTRACT

The HIV protease inhibitor nelfinavir is currently being analyzed for repurposing as an anticancer drug for many different cancers because it exerts manifold off-target protein interactions, finally resulting in cancer cell death. Xenosensing pregnane X receptor (PXR), which also participates in the control of cancer cell proliferation and apoptosis, was previously shown to be activated by nelfinavir; however, the exact molecular mechanism is still unknown. The present study addresses the effects of nelfinavir and its major and pharmacologically active metabolite nelfinavir hydroxy-tert-butylamide (M8) on PXR to elucidate the underlying molecular mechanism. Molecular docking suggested direct binding to the PXR ligand-binding domain, which was confirmed experimentally by limited proteolytic digestion and competitive ligand-binding assays. Concentration-response analyses using cellular transactivation assays identified nelfinavir and M8 as partial agonists with EC50 values of 0.9 and 7.3 µM and competitive antagonists of rifampin-dependent induction with IC50 values of 7.5 and 25.3 µM, respectively. Antagonism exclusively resulted from binding into the PXR ligand-binding pocket. Impaired coactivator recruitment by nelfinavir as compared with the full agonist rifampin proved to be the underlying mechanism of both effects on PXR. Physiologic relevance of nelfinavir-dependent modulation of PXR activity was investigated in respectively treated primary human hepatocytes, which showed differential induction of PXR target genes and antagonism of rifampin-induced ABCB1 and CYP3A4 gene expression. In conclusion, we elucidate here the molecular mechanism of nelfinavir interaction with PXR. It is hypothesized that modulation of PXR activity may impact the anticancer effects of nelfinavir. SIGNIFICANCE STATEMENT: Nelfinavir, which is being investigated for repurposing as an anticancer medication, is shown here to directly bind to human pregnane X receptor (PXR) and thereby act as a partial agonist and competitive antagonist. Its major metabolite nelfinavir hydroxy-tert-butylamide exerts the same effects, which are based on impaired coactivator recruitment. Nelfinavir anticancer activity may involve modulation of PXR, which itself is discussed as a therapeutic target in cancer therapy and for the reversal of chemoresistance.


Subject(s)
Hepatocytes/metabolism , Nelfinavir/analogs & derivatives , Nelfinavir/pharmacology , Pregnane X Receptor/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Binding Sites , Cytochrome P-450 CYP3A/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Nelfinavir/chemistry , Pregnane X Receptor/agonists , Pregnane X Receptor/antagonists & inhibitors , Pregnane X Receptor/chemistry , Primary Cell Culture
3.
J Mol Graph Model ; 103: 107803, 2021 03.
Article in English | MEDLINE | ID: mdl-33333424

ABSTRACT

In this study, the binding strength of 32 diastereomers of nelfinavir, a proposed drug for the treatment of COVID-19, was considered against main protease. Molecular docking was used to determine the most potent diastereomers. The top three diastereomers along with apo form of protein were then considered via molecular dynamics simulation and MM-GBSA method. During the simulation, the structural consideration of four proteins considered was carried out using RMSD, RMSF, Rg and hydrogen bond analysis tools. Our data demonstrated that the effect of nelfinavir RSRSR stereoisomer on protein stability and compactness is higher than the other. We also found from the hydrogen bond analysis that this important diastereomer form three hydrogen bonds with the residues of Glu166, Gly143 and Hie41. MM/GBSA analysis showed that the binding strength of RSRSR is more than other stereoisomers and that the main contributions to binding energy are vdW and electronic terms. The nelfinavir RSRSR stereoisomer introduced in this study may be effective in the treatment of COVID-19.


Subject(s)
Antiviral Agents/chemistry , Apoproteins/antagonists & inhibitors , Coronavirus 3C Proteases/antagonists & inhibitors , Nelfinavir/chemistry , Protease Inhibitors/chemistry , SARS-CoV-2/chemistry , Antiviral Agents/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Binding Sites , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Hydrogen Bonding , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Nelfinavir/metabolism , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , SARS-CoV-2/enzymology , Stereoisomerism , Thermodynamics
4.
Biomed Res Int ; 2020: 6237160, 2020.
Article in English | MEDLINE | ID: mdl-33102585

ABSTRACT

Coronaviruses have been reported previously due to their association with the severe acute respiratory syndrome (SARS). After SARS, these viruses were known to be causing Middle East respiratory syndrome (MERS) and caused 35% evanescence amid victims pursuing remedial care. Nowadays, beta coronaviruses, members of Coronaviridae, family order Nidovirales, have become subjects of great importance due to their latest pandemic originating from Wuhan, China. The virus named as human-SARS-like coronavirus-2 contains four structural as well as sixteen nonstructural proteins encoded by single-stranded ribonucleic acid of positive polarity. As there is no vaccine available to treat the infection caused by these viruses, there is a dire need for taking necessary steps against this virus. Herein, we have targeted two nonstructural proteins of SARS-CoV-2, namely, methyltransferase (nsp16) and helicase (nsp13), respectively, due to their substantial activity in viral pathogenesis. A total of 2035 compounds were analyzed for their pharmacokinetics and pharmacological properties. The screened 108 compounds were docked against both targeted proteins and were compared with previously reported known compounds. Compounds with high binding affinity were analyzed for their reactivity through DFT analysis, and binding was analyzed using molecular dynamics simulations. Through the analyses performed in this study, it is concluded that EryvarinM, Silydianin, Osajin, and Raddeanine can be considered potential inhibitors for MTase, while TomentodiplaconeB, Osajin, Sesquiterpene Glycoside, Rhamnetin, and Silydianin for helicase after these compounds are validated thoroughly using in vitro and in vivo protocols.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Phytochemicals/chemistry , Phytochemicals/pharmacology , SARS-CoV-2/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/pharmacology , Antimetabolites/chemistry , Antimetabolites/pharmacology , Antiviral Agents/chemistry , COVID-19/epidemiology , COVID-19/virology , China/epidemiology , Dioxolanes/chemistry , Dioxolanes/pharmacology , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacology , Humans , Methyltransferases/drug effects , Molecular Docking Simulation , Nelfinavir/chemistry , Nelfinavir/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Protein Conformation , RNA Helicases/drug effects , SARS-CoV-2/chemistry , SARS-CoV-2/isolation & purification , SARS-CoV-2/metabolism , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
5.
Pharm Res ; 37(7): 123, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32514688

ABSTRACT

PURPOSE: Nelfinavir (NFV), a FDA approved antiretroviral drug, has been reported to exhibit cancer cells growth inhibition and increased apoptosis. However, it requires a higher dose leading to toxicity, thus limiting its potential clinical translation. We aim to develop biodegradable (poly (lactic-co-glycolic acid)) PLGA nanoparticles of nelfinavir and determine their efficacy to treat non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: HIV protease inhibitor, NFV, was loaded into PLGA nanoparticles by double emulsion/solvent evaporation method; and nanoparticles were characterized for physicochemical characteristics including morphology and intracellular uptake. Their anti-cancer efficacy in NSCLC was assessed by in vitro assays including cytotoxicity, cellular migration, colony formation; and 3D spheroid culture mimicking in-vivo tumor microenvironment. Studies were also conducted to elucidate effects on molecular pathways including apoptosis, autophagy, and endoplasmic stress. RESULTS: NFV loaded PLGA nanoparticles (NPs) were found to have particle size: 191.1 ± 10.0 nm, zeta potential: -24.3 ± 0.9 mV, % drug loading: 2.5 ± 0.0%; and entrapment efficiency (EE): 30.1 ± 0.5%. NFV NP inhibited proliferation of NSCLC cells compared to NFV and exhibited significant IC50 reduction. From the caspase-dependent apoptosis assays and western blot studies (upregulation of ATF3), it was revealed that NFV NP significantly induced ER stress marker ATF3, cleaved PARP and further caused autophagy inhibition (LC3BII upregulation) leading to increased cellular death. In addition, NFV NP were found to be more efficacious in penetrating solid tumors in ex-vivo studies compared to plain NFV. CONCLUSIONS: Nelfinavir, a lead HIV protease inhibitor can be repositioned as a NSCLC therapeutic through nanoparticulate delivery. Given its ability to induce apoptosis and efficient tumor penetration capability, NFV loaded PLGA nanoparticulate systems provide a promising delivery system in NSCLC treatment.


Subject(s)
Anti-HIV Agents/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Nanocapsules/chemistry , Nanotechnology/methods , Nelfinavir/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Anti-HIV Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation , Drug Compounding/methods , Drug Liberation , Drug Repositioning , Drug Stability , Gene Expression Regulation/drug effects , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nelfinavir/pharmacology
6.
J Med Virol ; 92(10): 2087-2095, 2020 10.
Article in English | MEDLINE | ID: mdl-32374457

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS CoV-2) is the causative agent of the coronavirus disease-2019 (COVID-19) pandemic. Coronaviruses enter cells via fusion of the viral envelope with the plasma membrane and/or via fusion of the viral envelope with endosomal membranes after virion endocytosis. The spike (S) glycoprotein is a major determinant of virus infectivity. Herein, we show that the transient expression of the SARS CoV-2 S glycoprotein in Vero cells caused extensive cell fusion (formation of syncytia) in comparison to limited cell fusion caused by the SARS S glycoprotein. Both S glycoproteins were detected intracellularly and on transfected Vero cell surfaces. These results are in agreement with published pathology observations of extensive syncytia formation in lung tissues of patients with COVID-19. These results suggest that SARS CoV-2 is able to spread from cell-to-cell much more efficiently than SARS effectively avoiding extracellular neutralizing antibodies. A systematic screening of several drugs including cardiac glycosides and kinase inhibitors and inhibitors of human immunodeficiency virus (HIV) entry revealed that only the FDA-approved HIV protease inhibitor, nelfinavir mesylate (Viracept) drastically inhibited S-n- and S-o-mediated cell fusion with complete inhibition at a 10-µM concentration. In-silico docking experiments suggested the possibility that nelfinavir may bind inside the S trimer structure, proximal to the S2 amino terminus directly inhibiting S-n- and S-o-mediated membrane fusion. Also, it is possible that nelfinavir may act to inhibit S proteolytic processing within cells. These results warrant further investigations of the potential of nelfinavir mesylate to inhibit virus spread at early times after SARS CoV-2 symptoms appear.


Subject(s)
Anti-HIV Agents/pharmacology , Membrane Fusion/drug effects , Nelfinavir/pharmacology , SARS-CoV-2/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Animals , Anti-HIV Agents/chemistry , Binding Sites , Cell Fusion , Chlorocebus aethiops , Giant Cells/drug effects , Giant Cells/pathology , Giant Cells/virology , Humans , Molecular Docking Simulation , Nelfinavir/chemistry , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Virion/drug effects , Virion/pathogenicity , Virion/physiology , COVID-19 Drug Treatment
7.
Phys Chem Chem Phys ; 22(8): 4464-4480, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32057044

ABSTRACT

Infection by human immunodeficiency virus type 1 (HIV-1) not only destroys the immune system bringing about acquired immune deficiency syndrome (AIDS), but also induces serious neurological diseases including behavioral abnormalities, motor dysfunction, toxoplasmosis, and HIV-1 associated dementia. The emergence of HIV-1 multidrug-resistant mutants has become a major problem in the therapy of patients with HIV-1 infection. Focusing on the wild type (WT) and G48T/L89M mutated forms of HIV-1 protease (HIV-1 PR) in complex with amprenavir (APV), indinavir (IDV), ritonavir (RTV), and nelfinavir (NFV), we have investigated the conformational dynamics and the resistance mechanism due to the G48T/L89M mutations by conducting a series of molecular dynamics (MD) simulations and free energy (MM-PBSA and solvated interaction energy (SIE)) analyses. The simulation results indicate that alterations in the side-chains of G48T/L89M mutated residues cause the inner active site to increase in volume and induce more curling of the flap tips, which provide the main contributions to weaker binding of inhibitors to the HIV-1 PR. The results of energy analysis reveal that the decrease in van der Waals interactions of inhibitors with the mutated PR relative to the wild-type (WT) PR mostly drives the drug resistance of mutations toward these four inhibitors. The energy decomposition analysis further indicates that the drug resistance of mutations can be mainly attributed to the change in van der Waals and electrostatic energy of some key residues (around Ala28/Ala28' and Ile50/Ile50'). Our work can give significant guidance to design a new generation of anti-AIDS inhibitors targeting PR in the therapy of patients with HIV-1 infection.


Subject(s)
HIV Protease/metabolism , Molecular Dynamics Simulation , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Carbamates/chemistry , Carbamates/metabolism , Drug Resistance/drug effects , Drug Resistance/genetics , Furans , HIV Protease/genetics , Indinavir/chemistry , Indinavir/metabolism , Molecular Conformation , Mutation , Nelfinavir/chemistry , Nelfinavir/metabolism , Protein Binding , Ritonavir/chemistry , Ritonavir/metabolism , Sulfonamides/chemistry , Sulfonamides/metabolism
8.
AAPS PharmSciTech ; 19(8): 3584-3598, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30209788

ABSTRACT

Nelfinavir mesylate (NFV), a human immunodeficiency virus (HIV) protease inhibitor, is an integral component of highly active anti retro viral therapy (HAART) for management of AIDS. NFV possesses pH-dependent solubility and has low and variable bioavailability hampering its use in therapeutics. Lipid-based particulates have shown to improve solubility of poorly water soluble drugs and oral absorption, thereby aiding in improved bioavailability. The current study compares potential of vesicular and solid lipid nanocarriers of NFV with drug nanocrystallites and microvesicular systems like cochleates in improving bioavailability of NFV. The paper outlines investigation of systems using in vitro models like in vitro lipolysis, in vitro release, and permeation through cell lines to predict the in vivo potential of nanocarriers. Finally, in vivo pharmacokinetic study is reported which provided proof of concept in sync with results from in vitro studies. Graphical Abstract ᅟ.


Subject(s)
HIV Protease Inhibitors/chemistry , Lipids/chemistry , Nelfinavir/chemistry , Animals , Biological Availability , Caco-2 Cells , Female , Humans , Nelfinavir/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility
9.
J Mol Graph Model ; 75: 390-402, 2017 08.
Article in English | MEDLINE | ID: mdl-28645089

ABSTRACT

HIV-1 protease plays a crucial role in viral replication and maturation, which makes it one of the most attractive targets for anti-retroviral therapy. The majority of HIV infections in developing countries are due to non-B subtype. Subtype AE is spreading rapidly and infecting huge population worldwide. The mutations in the active site of subtype AE directly impair the interactions with the inhibitor. The non-active site mutations influence the binding of the inhibitor indirectly and their resistance mechanism is not well understood. It is important to design new effective inhibitors that combat drug resistance in subtype AE protease. In this work, we examined the effect of non active site mutations L10F, L10F/N88S and L90M with nelfinavir using molecular dynamics simulation and binding free energy calculations. The simulations suggested that the L10F and L10F/N88S mutants decrease the binding affinity of nelfinavir, whereas the L90M mutant increases the binding affinity. The formation of hydrogen bonds between nelfinavir and Asp30 is crucial for effective binding. The benzamide moiety of nelfinavir shows large positional deviation in L10F and L10F/N88S complexes and the L10F/N88S mutation changes the hydrogen bond between the side chain atoms of 30th residue and the 88th residue. Consequently the hydrogen bond interaction between Asp30 and nelfinavir are destroyed leading to drug resistance. Our present study shed light on the resistance mechanism of the strongly linked mutation L10F/N88S observed experimentally in AE subtype.


Subject(s)
Drug Resistance, Viral/genetics , HIV Protease/genetics , Molecular Dynamics Simulation , Mutation/genetics , Amino Acid Sequence , Biocatalysis , HIV Protease/chemistry , Humans , Hydrogen Bonding , Mutant Proteins/chemistry , Nelfinavir/chemistry , Nelfinavir/pharmacology , Sequence Alignment , Structural Homology, Protein , Thermodynamics
11.
AIDS ; 30(17): 2729-2730, 2016 11 13.
Article in English | MEDLINE | ID: mdl-27662548

ABSTRACT

Ethyl methyl sulfone contained in nelfinavir between 2007 and 2008 accidentally exposed embryos and fetuses to a powerful mutagen. We report data for 101 HIV-uninfected children exposed in utero included in the French prospective national cohort. The incidence of malformation was similar to that in the cohort as a whole with different drug exposures; no children had developed cancer after 9 years of follow-up.


Subject(s)
Congenital Abnormalities/epidemiology , Ethyl Methanesulfonate/analogs & derivatives , HIV Protease Inhibitors/administration & dosage , Maternal-Fetal Exchange , Mutagens/adverse effects , Nelfinavir/administration & dosage , Prenatal Exposure Delayed Effects/epidemiology , Adolescent , Child , Child, Preschool , Drug Contamination , Ethyl Methanesulfonate/administration & dosage , Ethyl Methanesulfonate/adverse effects , Female , Follow-Up Studies , France/epidemiology , HIV Protease Inhibitors/chemistry , Humans , Infant , Infant, Newborn , Male , Mutagens/administration & dosage , Nelfinavir/chemistry , Pregnancy , Prospective Studies
12.
EMBO Rep ; 17(10): 1471-1484, 2016 10.
Article in English | MEDLINE | ID: mdl-27572820

ABSTRACT

Activation of the elongation factor 2 kinase (eEF2K) leads to the phosphorylation and inhibition of the elongation factor eEF2, reducing mRNA translation rates. Emerging evidence indicates that the regulation of factors involved in protein synthesis may be critical for controlling diverse biological processes including cancer progression. Here we show that inhibitors of the HIV aspartyl protease (HIV-PIs), nelfinavir in particular, trigger a robust activation of eEF2K leading to the phosphorylation of eEF2. Beyond its anti-viral effects, nelfinavir has antitumoral activity and promotes cell death. We show that nelfinavir-resistant cells specifically evade eEF2 inhibition. Decreased cell viability induced by nelfinavir is impaired in cells lacking eEF2K. Moreover, nelfinavir-mediated anti-tumoral activity is severely compromised in eEF2K-deficient engrafted tumors in vivo Our findings imply that exacerbated activation of eEF2K is detrimental for tumor survival and describe a mechanism explaining the anti-tumoral properties of HIV-PIs.


Subject(s)
Elongation Factor 2 Kinase/metabolism , Neoplasms/metabolism , Neoplasms/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Drug Resistance/genetics , Elongation Factor 2 Kinase/genetics , Female , Gene Expression , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Nelfinavir/chemistry , Nelfinavir/pharmacology , Neoplasms/genetics , Peptide Elongation Factor 2/metabolism , Phosphorylation , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Tumor Burden
13.
Drug Des Devel Ther ; 10: 1837-46, 2016.
Article in English | MEDLINE | ID: mdl-27330277

ABSTRACT

OBJECTIVE: The standard treatment for cervical cancer in developed countries includes surgery and chemoradiation, with standard of care lagging in developing countries. Even in the former case, treatment frequently yields recalcitrant tumors and women succumb to disease. Here we examine the impact of nelfinavir, an off-patent viral protease inhibitor, which has shown promise as an antineoplastic agent. METHODS: We evaluated the morphological and proliferative effects of the autophagy-stressing drug nelfinavir in normal and cisplatin-resistant cervical cancer cells. Immunofluorescent validation of autophagy markers was performed and the impact of nelfinavir in an in vivo model of tumor growth was determined. RESULTS: Nelfinavir exhibits cytotoxicity against both cisplatin-sensitive and -resistant ME-180 human cervical cancer cells in vitro and in vivo. Immunoblotting and immunofluorescence showed an expression of the autophagy marker LC3-II in response to nelfinavir treatment. CONCLUSION: Nelfinavir, now available as an inexpensive generic orally dosed agent (Nelvir), is cytotoxic against cervical cancer cells. It acts by burdening the autophagy pathway to impair tumor cell survival and a modest induction of apoptosis. While further studies are needed to elucidate the optimal method of application of nelfinavir, it may represent an appealing global option for the treatment of cervical cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cisplatin/pharmacology , Nelfinavir/pharmacology , Uterine Cervical Neoplasms/pathology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cisplatin/chemistry , Female , Humans , Nelfinavir/chemistry
14.
Int J Pharm ; 501(1-2): 311-25, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26854426

ABSTRACT

Poor aqueous solubility and moderate permeability of Nelfinavir mesylate (NFM) leads to high variability in absorption after oral administration. To improve the solubility and bioavailability of NFM, the self microemulsifying drug delivery system (SMEDDS) was developed. For this purpose, Quality by design (QbD) approach employing D-optimal mixture design was used to prepare SMEDDS of NFM. Further, the software generated numerically optimized SMEDDS were developed by utilizing desirability function. Maisine 35-1, Tween 80, and Transcutol HP were identified as oil, surfactant, and co-surfactant that had best solubility for NFM. Ternary phase diagrams were plotted to identify the self-emulsification region. Dissolution of putative NFM in simulated fasted or fed small intestinal conditions, respectively, predicted that there is a positive food effect. However, NFM loaded SMEDDS showed absence of food effect with no significant difference in dissolution performance either in Fasted or fed state simulated intestinal fluid (FaSSIF or FeSSIF) biorelevent dissolution media. The prepared SMEDDS were thermodynamically stable with droplet size (121 nm), poly dispersity index (PDI) (0.198) and emulsification time (<1 min). Transmission electron microscopy (TEM) analysis confirmed the spherical shape of the reconstituted SMEDDS droplets. The ex vivo performance revealed 4.57 fold enhancement in the apparent permeability of NFM as compared to NFM suspension. The animal pharmacokinetic analysis in New Zealand strain rabbits indicated food effect on pure NFM suspension. However, absence of food effect and 3.5-3.6 fold enhancement in the oral bioavailability was observed when NFM was formulated into SMEDDS. Thus, it could be envisaged that development of SMEDDS formulation of NFM could be one of the best alternative to enhance oral bioavailability of NFM.


Subject(s)
Drug Delivery Systems , Nelfinavir/administration & dosage , Animals , Biological Availability , Chemistry, Pharmaceutical , Emulsions , Ethylene Glycols/administration & dosage , Ethylene Glycols/chemistry , Fasting/metabolism , Food-Drug Interactions , Gastrointestinal Tract/metabolism , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacokinetics , Intestinal Absorption/drug effects , Linoleic Acids/administration & dosage , Linoleic Acids/chemistry , Male , Nelfinavir/blood , Nelfinavir/chemistry , Nelfinavir/pharmacokinetics , Permeability/drug effects , Polysorbates/administration & dosage , Polysorbates/chemistry , Rabbits , Rats , Solubility , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry
15.
Curr Top Med Chem ; 16(19): 2156-68, 2016.
Article in English | MEDLINE | ID: mdl-26881715

ABSTRACT

Increased investments and development of new technologies in drug discovery have barely improved the outcome of medicinal entities in the drug discovery market from a long time. Minimal success rates of drug approvals, poor safety profiles, and long development processes are some of many hurdles encountered in the drug discovery field. Therefore, drug repurposing can provide an alternative approach to meet the demands of the new, potent and safe anti-cancer agents in terms of both economic cost and time efficiency. The common molecular pathways of different diseases and secondary indications of most of the approved drugs, and advances in genomics, informatics and biology, as well as the availability of approved or safe drug libraries can certainly provide an improved and efficient way of screening safer drugs for new indications. Promising results of drug repurposing in different therapeutic areas have encouraged the scientific community to discover new drugs for different diseases using this methodology. Herein, we provide a general overview of structurally and functionally diverse approved drugs that have been repurposed as anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning/methods , Antineoplastic Agents/chemistry , Aspirin/chemistry , Aspirin/pharmacology , Digoxin/chemistry , Digoxin/pharmacology , Diphosphonates/chemistry , Diphosphonates/pharmacology , Drug Approval , Drug Discovery , Humans , Itraconazole/chemistry , Itraconazole/pharmacology , Metformin/chemistry , Metformin/pharmacology , Nelfinavir/chemistry , Nelfinavir/pharmacology , Thalidomide/chemistry , Thalidomide/pharmacology
16.
PLoS One ; 11(1): e0146529, 2016.
Article in English | MEDLINE | ID: mdl-26741368

ABSTRACT

Cytochrome P450 3A4 (CYP3A4) is the major drug metabolic enzyme, and is involved in the metabolism of antiretroviral drugs, especially protease inhibitors (PIs). This study was undertaken to examine the effect of methamphetamine on the binding and metabolism of PIs with CYP3A4. We showed that methamphetamine exhibits a type I spectral change upon binding to CYP3A4 with δAmax and KD of 0.016±0.001 and 204±18 µM, respectively. Methamphetamine-CYP3A4 docking showed that methamphetamine binds to the heme of CYP3A4 in two modes, both leading to N-demethylation. We then studied the effect of methamphetamine binding on PIs with CYP3A4. Our results showed that methamphetamine alters spectral binding of nelfinavir but not the other type I PIs (lopinavir, atazanavir, tipranavir). The change in spectral binding for nelfinavir was observed at both δAmax (0.004±0.0003 vs. 0.0068±0.0001) and KD (1.42±0.36 vs.2.93±0.08 µM) levels. We further tested effect of methamphetamine on binding of 2 type II PIs; ritonavir and indinavir. Our results showed that methamphetamine alters the ritonavir binding to CYP3A4 by decreasing both the δAmax (0.0038±0.0003 vs. 0.0055±0.0003) and KD (0.043±0.0001 vs. 0.065±0.001 nM), while indinavir showed only reduced KD in presence of methamphetamine (0.086±0.01 vs. 0.174±0.03 nM). Furthermore, LC-MS/MS studies in high CYP3A4 human liver microsomes showed a decrease in the formation of hydroxy ritonavir in the presence of methamphetamine. Finally, CYP3A4 docking with lopinavir and ritonavir in the absence and presence of methamphetamine showed that methamphetamine alters the docking of ritonavir, which is consistent with the results obtained from spectral binding and metabolism studies. Overall, our results demonstrated differential effects of methamphetamine on the binding and metabolism of PIs with CYP3A4. These findings have clinical implication in terms of drug dose adjustment of antiretroviral medication, especially with ritonavir-boosted antiretroviral therapy, in HIV-1-infected individuals who abuse methamphetamine.


Subject(s)
Cytochrome P-450 CYP3A/chemistry , HIV Infections/drug therapy , HIV Protease Inhibitors/chemistry , Methamphetamine/chemistry , Amphetamine-Related Disorders/enzymology , Atazanavir Sulfate/chemistry , Atazanavir Sulfate/metabolism , Atazanavir Sulfate/pharmacology , Catalytic Domain , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , HIV Protease Inhibitors/pharmacology , Humans , Inactivation, Metabolic , Lopinavir/chemistry , Lopinavir/metabolism , Lopinavir/pharmacology , Methamphetamine/pharmacology , Microsomes, Liver/enzymology , Molecular Docking Simulation , Nelfinavir/chemistry , Nelfinavir/metabolism , Nelfinavir/pharmacology , Protein Binding , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology , Pyrones/chemistry , Pyrones/metabolism , Pyrones/pharmacology , Sulfonamides
17.
Drug Des Devel Ther ; 9: 6055-65, 2015.
Article in English | MEDLINE | ID: mdl-26622167

ABSTRACT

Based on experimental data, the anticancer activity of nelfinavir (NFV), a US Food and Drug Administration (FDA)-approved HIV-1 protease inhibitor (PI), was reported. Nevertheless, the mechanism of action of NFV is yet to be verified. It was hypothesized that the anticancer activity of NFV is due to its inhibitory effect on heat shock protein 90 (Hsp90), a promising target for anticancer therapy. Such findings prompted us to investigate the potential anticancer activity of all other FDA-approved HIV-1 PIs against human Hsp90. To accomplish this, "loop docking" - an enhanced in-house developed molecular docking approach - followed by molecular dynamic simulations and postdynamic analyses were performed to elaborate on the binding mechanism and relative binding affinities of nine FDA-approved HIV-1 PIs against human Hsp90. Due to the lack of the X-ray crystal structure of human Hsp90, homology modeling was performed to create its 3D structure for subsequent simulations. Results showed that NFV has better binding affinity (ΔG =-9.2 kcal/mol) when compared with other PIs: this is in a reasonable accordance with the experimental data (IC50 3.1 µM). Indinavir, saquinavir, and ritonavir have close binding affinity to NFV (ΔG =-9.0, -8.6, and -8.5 kcal/mol, respectively). Per-residue interaction energy decomposition analysis showed that hydrophobic interaction (most importantly with Val534 and Met602) played the most predominant role in drug binding. To further validate the docking outcome, 5 ns molecular dynamic simulations were performed in order to assess the stability of the docked complexes. To our knowledge, this is the first account of detailed computational investigations aimed to investigate the potential anticancer activity and the binding mechanism of the FDA-approved HIV PIs binding to human Hsp90. Information gained from this study should also provide a route map toward the design, optimization, and further experimental investigation of potential derivatives of PIs to treat HER2+ breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning , HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , HIV-1/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Nelfinavir/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Drug Design , HIV Protease/chemistry , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/metabolism , HIV-1/enzymology , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , Nelfinavir/chemistry , Nelfinavir/metabolism , Protein Binding , Protein Conformation , Reproducibility of Results , Structure-Activity Relationship
18.
BMC Bioinformatics ; 16 Suppl 19: S10, 2015.
Article in English | MEDLINE | ID: mdl-26695135

ABSTRACT

BACKGROUND: The human immunodeficiency virus (HIV-1) is a retrovirus causing acquired immunodeficiency syndrome (AIDS), which has become a serious problem across the world and has no cure reported to date. Human immunodeficiency virus (HIV-1) protease is an attractive target for antiviral treatment and a number of therapeutically useful inhibitors have been designed against it. The emergence of drug resistant mutants of HIV-1 poses a serious problem for conventional therapies that have been used so far. Until now, thirteen protease inhibitors (PIs), major mutation sites and many secondary mutations have been listed in the HIV Drug Resistance Database. In this study, we have studied the effect of the V77I mutation in HIV-PR along with the co-occurring mutations L33F and K20T through multi-nanosecond molecular dynamics simulations. V77I is known to cause Nelfinavir (NFV) resistance in the subtype B population of HIV-1 protease. We have for the first time reported the effect of this clinically relevant mutation on the binding of Nelfinavir and the conformational flexibility of the protease. RESULTS: Two HIV-PR mutants have been considered in this study - the Double Mutant Protease (DBM) V77I-L33F and Triple Mutant Protease (TPM) V77I-K20T-L33F. The molecular dynamics simulation studies were carried out and the RMSD trajectories of the unliganded wild type and mutated protease were found to be stable. The binding affinity of NFV with wild type HIV-PR was very high with a Glide XP docking score of -9.3 Kcal/mol. NFV showed decreased affinity towards DBM with a docking score of -8.0 Kcal/mol, whereas its affinity increased towards TPM (Glide XP score: -10.3). Prime/MM-GBSA binding free energy of the wild type, DBM and TPM HIV-PR docked structures were calculated as -38.9, -11.1 and -42.6 Kcal/mol respectively. The binding site cavity volumes of wild type, DBM and TPM protease were 1186.1, 1375.5 and 1042.5 Å3 respectively. CONCLUSION: In this study, we have studied the structural roles of the two HIV-PR mutations by conducting molecular dynamics simulation studies of the wild type and mutant HIV-1 PRs. The present study proposes that DBM protease showed greater flexibility and the flap separation was greater with respect to the wild type protease. The cavity size of the MD-stabilized DBM was also found to be increased, which may be responsible for the decreased interaction of Nelfinavir with the cavity residues, thus explaining the decreased binding affinity. On the other hand, the binding affinity of NFV for TPM was found to be enhanced, accounted for by the decrease in cavity size of the mutant which facilitated strong interactions with the flap residues. The flap separation of TPM was less than the wild type protease and the decreased cavity size may be responsible for its lower resistance, and hence, may be the reason for its lower clinical relevance.


Subject(s)
Drug Resistance, Viral/genetics , HIV Protease/genetics , HIV-1/enzymology , HIV-1/genetics , Mutation/genetics , Nelfinavir/chemistry , Nelfinavir/pharmacology , Binding Sites/genetics , Catalytic Domain , HIV Infections/genetics , HIV Protease Inhibitors/pharmacology , HIV-1/isolation & purification , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Thermodynamics
19.
J Photochem Photobiol B ; 151: 194-200, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26310356

ABSTRACT

Understanding the interaction of small molecules with DNA has become an active research area at the interface between biology and chemistry. In the present work, we investigated the mode of interaction of nelfinavir (NFV) with herring sperm DNA (hs DNA) under physiological conditions using various biophysical techniques. Analysis of UV-absorption and fluorescence spectra indicates the formation of complex between NFV and hs DNA. According to the fluorescence results, the binding constant (K) between NFV and hs DNA was found to be 3.30 × 10(4)LM(-1). The calculated thermodynamic parameters (ΔH° and ΔS°) suggested that hydrogen bonding plays a major role in binding between them. Phosphate group binding studies revealed that there was no electrostatic interactions occurred between NFV and hs DNA. Circular dichroism (CD) and DNA melting curve were employed to measure the conformational change of hs DNA in the presence of NFV, which verified the minor groove binding mode. These results were further supported by viscosity measurements and competitive displacement assay study using Hoechst 33258. According to the sequence specificity experiments, NFV binds to A-T rich region of hs DNA.


Subject(s)
DNA/metabolism , Ethidium/metabolism , Nelfinavir/chemistry , Nelfinavir/metabolism , Animals , Binding Sites , Binding, Competitive , Circular Dichroism , DNA/chemistry , Ethidium/chemistry , Fishes/genetics , Fluorescence , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HIV Protease Inhibitors/metabolism , Hydrogen Bonding , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Static Electricity , Thermodynamics , Viscosity
20.
Sci Rep ; 5: 9698, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25880275

ABSTRACT

Nelfinavir and its analogs inhibit proliferation and induce apoptosis of castration-resistant prostate cancer through inhibition of site-2 protease (S2P) activity, which leads to suppression of regulated intramembrane proteolysis. Western blotting in nelfinavir and its analog treated cells confirms accumulation of precursor SREBP-1 and ATF6. Nelfinavir and its analogs inhibit human homolog M. jannaschii S2P cleavage of an artificial protein substrate CED-9 in an in vitro proteolysis assay in a dose-dependent manner. Nelfinavir and its analogs are more potent inhibitors of S2P cleavage activity than 1,10-phenanthroline, a metalloprotease-specific inhibitor. Further, cluster analysis of gene expression from treated DU145 and PC3 cell lines demonstrate a close similarity of nelfinavir, its analogs, and 1,10-phenanthroline. These results show nelfinavir and its analogs inhibit castration-resistant prostate cancer proliferation by blocking regulated intramembrane proteolysis through suppression of S2P cleavage activity. This leads to accumulation of precursor SREBP-1 and ATF6, and development of insufficient reserves of their transcriptionally-active forms. The present results validate S2P and regulated intramembrane proteolysis as novel therapeutic targets for castration-resistant prostate cancer therapeutics. A clinical trial of nelfinavir or its analogs should be developed for castration-resistant prostate cancer.


Subject(s)
Metalloendopeptidases/antagonists & inhibitors , Nelfinavir/chemistry , Activating Transcription Factor 6/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Humans , Male , Metalloendopeptidases/metabolism , Methanocaldococcaceae/enzymology , Nelfinavir/therapeutic use , Nelfinavir/toxicity , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Precursors/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA/analysis , Sequence Analysis, RNA , Sterol Regulatory Element Binding Protein 1/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...