Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Bioorg Chem ; 148: 107495, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805850

ABSTRACT

Targeting Ribonuclease H (RNase H) has been considered a viable strategy for HIV therapy. In this study, a series of novel thiazolo[3, 2-a]pyrimidine derivatives were firstly designed and synthesized as potential inhibitors of HIV-1 RNase H. Among these compounds, A28 exhibited the most potent inhibition against HIV-1 RNase H with an IC50 value of 4.14 µM, which was about 5-fold increase in potency than the hit compound A1 (IC50 = 21.49 µM). To gain deeper insights into the structure-activity relationship (SAR), a CoMFA model was constructed to yield reasonable statistical results (q2 = 0.658 and R2 = 0.969). Results from magnesium ion chelation experiments and molecular docking studies revealed that these thiazolopyrimidine inhibitors may exert their inhibitory activity by binding to an allosteric site on RNase H at the interface between subunits p51 and p66. Furthermore, this analog demonstrated favorable physicochemical properties. Our findings provide valuable groundwork for further development of allosteric inhibitors targeting HIV-1 RNase H.


Subject(s)
Drug Design , HIV-1 , Molecular Docking Simulation , Pyrimidines , Structure-Activity Relationship , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , HIV-1/drug effects , HIV-1/enzymology , Humans , Thiazoles/chemistry , Thiazoles/pharmacology , Thiazoles/chemical synthesis , Molecular Structure , Anti-HIV Agents/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/metabolism
2.
J Mol Biol ; 434(7): 167507, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35217069

ABSTRACT

In retroviruses, strand displacement DNA-dependent DNA polymerization catalyzed by the viral reverse transcriptase (RT) is required to synthesize double-stranded proviral DNA. In addition, strand displacement during RNA-dependent DNA synthesis is critical to generate high-quality cDNA for use in molecular biology and biotechnology. In this work, we show that the loss of RNase H activity due to inactivating mutations in HIV-1 RT (e.g. D443N or E478Q) has no significant effect on strand displacement while copying DNA templates, but has a large impact on DNA polymerization in reactions carried out with RNA templates. Similar effects were observed with ß-thujaplicinol and other RNase H active site inhibitors, including compounds with dual activity (i.e., characterized also as inhibitors of HIV-1 integrase and/or the RT DNA polymerase). Among them, dual inhibitors of HIV-1 RT DNA polymerase/RNase H activities, containing a 7-hydroxy-6-nitro-2H-chromen-2-one pharmacophore were found to be very potent and effective strand displacement inhibitors in RNA-dependent DNA polymerization reactions. These findings might be helpful in the development of transcriptomics technologies to obtain more uniform read coverages when copying long RNAs and for the construction of more representative libraries avoiding biases towards 5' and 3' ends, while providing valuable information for the development of novel antiretroviral agents.


Subject(s)
DNA, Viral , HIV Reverse Transcriptase , Ribonuclease H, Human Immunodeficiency Virus , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/pharmacology , DNA, Viral/biosynthesis , Drug Development , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Tropolone/analogs & derivatives , Tropolone/pharmacology
3.
Eur J Med Chem ; 225: 113769, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34403976

ABSTRACT

Despite significant advances in antiretroviral therapy, acquired immunodeficiency syndrome remains as one of the leading causes of death worldwide. New antiretroviral drugs combined with updated treatment strategies are needed to improve convenience, tolerability, safety, and antiviral efficacy of available therapies. In this work, a focused library of coumarin derivatives was exploited by cell phenotypic screening to discover novel inhibitors of HIV-1 replication. Five compounds (DW-3, DW-4, DW-11, DW-25 and DW-31) showed moderate activity against wild-type and drug-resistant strains of HIV-1 (IIIB and RES056). Four of those molecules were identified as inhibitors of the viral RT-associated RNase H. Structural modification of the most potent DW-3 and DW-4 led to the discovery of compound 8a. This molecule showed increased potency against wild-type HIV-1 strain (EC50 = 3.94 ± 0.22 µM) and retained activity against a panel of mutant strains, showing EC50 values ranging from 5.62 µM to 202 µM. In enzymatic assays, 8a was found to inhibit the viral RNase H with an IC50 of 12.3 µM. Molecular docking studies revealed that 8a could adopt a binding mode similar to that previously reported for other active site HIV-1 RNase H inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , Coumarins/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Discovery , HIV Reverse Transcriptase/metabolism , HIV-1/metabolism , Humans , Microbial Sensitivity Tests , Molecular Structure , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
4.
J Med Chem ; 64(12): 8579-8598, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34106711

ABSTRACT

Novel anti-HIV agents are still needed to overcome resistance issues, in particular inhibitors acting against novel viral targets. The ribonuclease H (RNase H) function of the reverse transcriptase (RT) represents a validated and promising target, and no inhibitor has reached the clinical pipeline yet. Here, we present rationally designed non-diketo acid selective RNase H inhibitors (RHIs) based on the quinolinone scaffold starting from former dual integrase (IN)/RNase H quinolinonyl diketo acids. Several derivatives were synthesized and tested against RNase H and viral replication and found active at micromolar concentrations. Docking studies within the RNase H catalytic site, coupled with site-directed mutagenesis, and Mg2+ titration experiments demonstrated that our compounds coordinate the Mg2+ cofactor and interact with amino acids of the RNase H domain that are highly conserved among naïve and treatment-experienced patients. In general, the new inhibitors influenced also the polymerase activity of RT but were selective against RNase H vs the IN enzyme.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/enzymology , Quinolones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/metabolism , HeLa Cells , Humans , Magnesium/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Protein Binding , Quinolones/chemical synthesis , Quinolones/metabolism , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/metabolism , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Virus Replication/drug effects
5.
Arch Pharm (Weinheim) ; 354(9): e2100123, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34008218

ABSTRACT

The bioactive components of Garcinia indica, garcinol (camboginol), and isogarcinol (cambogin), are suitable drug candidates for the treatment of various human diseases. HIV-1-RNase H assay was used to study the RNase H inhibition by garcinol and isogarcinol. Docking of garcinol into the active site of the enzyme was carried out to rationalize the difference in activities between the two compounds. Garcinol showed higher HIV-1-RNase H inhibition than the known inhibitor RDS1759 and retained full potency against the RNase H of a drug-resistant HIV-1 reverse transcriptase form. Isogarcinol was distinctly less active than garcinol, indicating the importance of the enolizable ß-diketone moiety of garcinol for anti-RNase H activity. Docking calculations confirmed these findings and suggested this moiety to be involved in the chelation of metal ions of the active site. On the basis of its HIV-1 reverse transcriptase-associated RNase H inhibitory activity, garcinol is worth being further explored concerning its potential as a cost-effective treatment for HIV patients.


Subject(s)
Garcinia/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Terpenes/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , HIV-1/enzymology , Molecular Docking Simulation , Reverse Transcriptase Inhibitors/isolation & purification , Terpenes/isolation & purification
6.
J Enzyme Inhib Med Chem ; 35(1): 1953-1963, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33143469

ABSTRACT

Bioisosteric replacement and scaffold hopping are powerful strategies in drug design useful for rationally modifying a hit compound towards novel lead therapeutic agents. Recently, we reported a series of thienopyrimidinones that compromise dynamics at the p66/p51 HIV-1 reverse transcriptase (RT)-associated Ribonuclease H (RNase H) dimer interface, thereby allosterically interrupting catalysis by altering the active site geometry. Although they exhibited good submicromolar activity, the isosteric replacement of the thiophene ring, a potential toxicophore, is warranted. Thus, in this article, the most active 2-(3,4-dihydroxyphenyl)-5,6-dimethylthieno[2,3-d]pyrimidin-4(3H)-one 1 was selected as the hit scaffold and several isosteric substitutions of the thiophene ring were performed. A novel series of highly active RNase H allosteric quinazolinone inhibitors was thus obtained. To determine their target selectivity, they were tested against RT-associated RNA-dependent DNA polymerase (RDDP) and integrase (IN). Interestingly, none of the compounds were particularly active on (RDDP) but many displayed micromolar to submicromolar activity against IN.


Subject(s)
Anti-HIV Agents/chemical synthesis , HIV Reverse Transcriptase/metabolism , Pyrimidinones/chemistry , Quinazolinones/chemical synthesis , Reverse Transcriptase Inhibitors/chemical synthesis , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/pharmacology , Catalytic Domain , Drug Design , Humans , Models, Molecular , Protein Binding , Protein Multimerization , Quinazolinones/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship , Thiophenes/chemistry
7.
Viruses ; 12(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32640577

ABSTRACT

HIV-1 infection requires life-long treatment and with 2.1 million new infections/year, faces the challenge of an increased rate of transmitted drug-resistant mutations. Therefore, a constant and timely effort is needed to identify new HIV-1 inhibitors active against drug-resistant variants. The ribonuclease H (RNase H) activity of HIV-1 reverse transcriptase (RT) is a very promising target, but to date, still lacks an efficient inhibitor. Here, we characterize the mode of action of N'-(2-hydroxy-benzylidene)-3,4,5-trihydroxybenzoylhydrazone (compound 13), an N-acylhydrazone derivative that inhibited viral replication (EC50 = 10 µM), while retaining full potency against the NNRTI-resistant double mutant K103N-Y181C virus. Time-of-addition and biochemical assays showed that compound 13 targeted the reverse-transcription step in cell-based assays and inhibited the RT-associated RNase H function, being >20-fold less potent against the RT polymerase activity. Docking calculations revealed that compound 13 binds within the RNase H domain in a position different from other selective RNase H inhibitors; site-directed mutagenesis studies revealed interactions with conserved amino acid within the RNase H domain, suggesting that compound 13 can be taken as starting point to generate a new series of more potent RNase H selective inhibitors active against circulating drug-resistant variants.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV-1/drug effects , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/pharmacology , Binding Sites , Drug Resistance, Viral , HIV-1/enzymology , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Mutagenesis, Site-Directed , Ribonuclease H
8.
Acc Chem Res ; 53(1): 218-230, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31880912

ABSTRACT

Human immunodeficiency virus (HIV) reverse transcriptase (RT) contains two distinct functional domains: a DNA polymerase (pol) domain and a ribonuclease H (RNase H) domain, both of which are required for viral genome replication. Over the last 3 decades, RT has been at the forefront of HIV drug discovery efforts with numerous nucleoside reverse transcriptase inhibitors (NRTIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) approved by the FDA. However, all these RT inhibitors target only the pol function, and inhibitors of RT-associated RNase H have yet to enter the development pipeline, which in itself manifests both the opportunity and challenges of targeting RNase H: if developed, RT RNase H inhibitors would represent a mechanistically novel class of HIV drugs that can be particularly valuable in treating HIV strains resistant to current drugs. The challenges include (1) the difficulty in selectively targeting RT RNase H over RT pol due to their close interplay both spatially and temporally and over HIV-1 integrase strand transfer (INST) activity because of their active site similarities; (2) to a larger extent, the inability of active site inhibitors to confer significant antiviral effect, presumably due to a steep substrate barrier by which the pre-existing substrate prevents access of small molecules to the active site. As a result, previously reported RT RNase H inhibitors typically lacked target specificity and significant antiviral potency. Achieving meaningful antiviral activity via active site targeting likely entails selective and ultrapotent RNase H inhibition to allow small molecules to cut into the dominance of substrates. Based on a pharmacophore model informed by prior work, we designed and redesigned a few metal-chelating chemotypes, such as 2-hydroxyisoquinolinedione (HID), hydroxypyridonecarboxylic acid (HPCA), 3-hydroxypyrimidine-2,4-dione (HPD), and N-hydroxythienopyrimidine-2,4-dione (HTPD). Analogues of these chemotypes generally exhibited improved potency and selectivity inhibiting RT RNase H over the best previous compounds and further validated the pharmacophore model. Extended structure-activity relationship (SAR) on the HPD inhibitor type by mainly altering the linkage generated a few subtypes showing exceptional potency (single-digit nanomolar) and excellent selectivity over the inhibition of RT pol and INST. In parallel, a structure-based approach also allowed us to design a unique double-winged HPD subtype to potently and selectively inhibit RT RNase H and effectively compete against the RNA/DNA substrate. Significantly, all potent HPD subtypes consistently inhibited HIV-1 in the cell culture, suggesting that carefully designed active site RNase H inhibitors with ultrapotency could partially overcome the barrier to antiviral phenotype. Overall, in addition to identifying our own inhibitor types, our medicinal chemistry efforts demonstrated the value of pharmacophore and structure-based approaches in designing active side-directed RNase H inhibitors and could provide a viable path to validating RNase H as a novel antiviral target.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , Humans , Models, Molecular , Molecular Structure , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism
9.
Antiviral Res ; 174: 104671, 2020 02.
Article in English | MEDLINE | ID: mdl-31812637

ABSTRACT

The management of Human Immunodeficiency Virus type 1 (HIV-1) infection requires life-long treatment that is associated with chronic toxicity and possible selection of drug-resistant strains. A new opportunity for drug intervention is offered by antivirals that act as allosteric inhibitors targeting two viral functions (dual inhibitors). In this work, we investigated the effects of 5,6-dihydroxyindole-2-carboxylic acid (DHICA) derivatives on both HIV-1 Integrase (IN) and Reverse Transcriptase associated Ribonuclease H (RNase H) activities. Among the tested compounds, the dihydroxyindole-carboxamide 5 was able to inhibit in the low micromolar range (1-18 µM) multiple functions of IN, including functional IN-IN interactions, IN-LEDGF/p75 binding and IN catalytic activity. Docking and site-directed mutagenesis studies have suggested that compound 5 binds to a previously described HIV-1 IN allosteric pocket. These observations indicate that 5 is structurally and mechanistically distinct from the published allosteric HIV-1 IN inhibitors. Moreover, compound 5 also inhibited HIV-1 RNase H function, classifying this molecule as a dual HIV-1 IN and RNase H inhibitor able to impair the HIV-1 virus replication in cell culture. Overall, we identified a new scaffold as a suitable platform for the development of novel dual HIV-1 inhibitors.


Subject(s)
Carboxylic Acids/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Carboxylic Acids/chemistry , Cell Line , Drug Discovery , HIV Infections/virology , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
10.
ACS Infect Dis ; 5(11): 1963-1974, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31577424

ABSTRACT

The ribonuclease H (RNH) activity of HIV-1 reverse transcriptase (RT) is essential for viral replication and can be a target for drug development. Yet, no RNH inhibitor to date has substantial antiviral activity to allow advancement into clinical development. Herein, we describe our characterization of the detailed binding mechanisms of RNH active-site inhibitors, YLC2-155 and ZW566, that bind to the RNH domain through divalent metal ions, using NMR, molecular docking, and quantum mechanical calculations. In the presence of Mg2+, NMR spectra of RNH exhibited split (two) resonances for some residues upon inhibitor binding, suggesting two binding modes, an observation consistent with the docking results. The relative populations of the two binding conformers were independent of inhibitor or Mg2+ concentration, with one conformation consistently more favored. In our docking study, one distinctive pose of ZW566 showed more interactions with surrounding residues of RNH compared to the analogous binding pose of YLC2-155. Inhibitor titration experiments revealed a lower dissociation constant for ZW566 compared to YLC2-155, in agreement with its higher inhibitory activity. Mg2+ titration data also indicated a stronger dependence on Mg2+ for the RNH interaction with ZW566 compared to YLC2-155. Combined docking and quantum mechanical calculation results suggest that stronger metal coordination as well as more protein-inhibitor interactions may account for the higher binding affinity of ZW566. These findings support the idea that strategies for the development of potent competitive active site RNH inhibitors should take into account not only metal-inhibitor coordination but also protein-inhibitor interaction and conformational selectivity.


Subject(s)
Anti-HIV Agents/chemistry , Enzyme Inhibitors/chemistry , HIV Infections/virology , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Anti-HIV Agents/metabolism , Catalytic Domain , Enzyme Inhibitors/metabolism , HIV-1/drug effects , HIV-1/genetics , Humans , Isoquinolines/chemistry , Isoquinolines/metabolism , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Molecular Docking Simulation , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism
11.
Eur J Med Chem ; 182: 111617, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31442684

ABSTRACT

A number of compounds targeting different processes of the Human Immunodeficiency Virus type 1 (HIV-1) life cycle have been developed in the continuing fight against AIDS. Coumarin-based molecules already proved to act as HIV-1 Protease (PR) or Integrase (IN) inhibitors and also to target HIV-1 reverse transcriptase (RT), blocking the DNA-dependent DNA-polymerase activity or the RNA-dependent DNA-polymerase activity working as common NNRTIs. In the present study, with the aim to exploit a coumarin-based scaffold to achieve the inhibition of multiple viral coded enzymatic functions, novel 4-hydroxy-2H, 5H-pyrano (3, 2-c) chromene-2, 5-dione derivatives were synthesized. The modeling studies calculated the theoretical binding affinity of the synthesized compounds on both HIV-1 IN and RT-associated Ribonuclease H (RNase H) active sites, which was confirmed by biological assays. Our results provide a basis for the identification of dual HIV-1 IN and RT RNase H inhibitors compounds.


Subject(s)
Anti-HIV Agents/pharmacology , Coumarins/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , Microbial Sensitivity Tests , Molecular Structure , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
12.
Bioorg Med Chem ; 27(17): 3836-3845, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31324562

ABSTRACT

A novel series of 3-hydroxyquinazoline-2,4(1H,3H)-diones derivatives has been designed and synthesized. Their biochemical characterization revealed that most of the compounds were effective inhibitors of HIV-1 RNase H activity at sub to low micromolar concentrations. Among them, II-4 was the most potent in enzymatic assays, showing an IC50 value of 0.41 ±â€¯0.13 µM, almost five times lower than the IC50 obtained with ß-thujaplicinol. In addition, II-4 was also effective in inhibiting HIV-1 IN strand transfer activity (IC50 = 0.85 ±â€¯0.18 µM) but less potent than raltegravir (IC50 = 71 ±â€¯14 nM). Despite its relatively low cytotoxicity, the efficiency of II-4 in cell culture was limited by its poor membrane permeability. Nevertheless, structure-activity relationships and molecular modeling studies confirmed the importance of tested 3-hydroxyquinazoline-2,4(1H,3H)-diones as useful leads for further optimization.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , HIV Integrase/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , Quinazolinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Caco-2 Cells , Cell Line , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-2/drug effects , Humans , Models, Molecular , Molecular Structure , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
13.
Eur J Med Chem ; 166: 390-399, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30739822

ABSTRACT

The pharmacophore of active site inhibitors of human immunodeficiency virus (HIV) reverse transcriptase (RT)-associated RNase H typically entails a flexible linker connecting the chelating core and the hydrophobic aromatics. We report herein that novel 3-hydroxypyrimidine-2,4-dione (HPD) subtypes with a nonflexible C-6 carbonyl linkage exhibited potent and selective biochemical inhibitory profiles with strong RNase H inhibition at low nM, weak to moderate integrase strand transfer (INST) inhibition at low µM, and no to marginal RT polymerase (pol) inhibition up to 10 µM. A few analogues also demonstrated significant antiviral activity without cytotoxicity. The overall inhibitory profile is comparable to or better than that of previous HPD subtypes with a flexible C-6 linker, suggesting that the nonflexible carbonyl linker can be tolerated in the design of novel HIV RNase H active site inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Catalytic Domain , Drug Design , Enzyme Inhibitors/metabolism , HIV-1/drug effects , Inhibitory Concentration 50 , Molecular Docking Simulation , Pyrimidinones/metabolism , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism
14.
J Enzyme Inhib Med Chem ; 34(1): 55-74, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30362381

ABSTRACT

The paper focussed on a step-by-step structural modification of a cycloheptathiophene-3-carboxamide derivative recently identified by us as reverse transcriptase (RT)-associated ribonuclease H (RNase H) inhibitor. In particular, its conversion to a 2-aryl-cycloheptathienoozaxinone derivative and the successive thorough exploration of both 2-aromatic and cycloheptathieno moieties led to identify oxazinone-based compounds as new anti-RNase H chemotypes. The presence of the catechol moiety at the C-2 position of the scaffold emerged as critical to achieve potent anti-RNase H activity, which also encompassed anti-RNA dependent DNA polymerase (RDDP) activity for the tricyclic derivatives. Benzothienooxazinone derivative 22 resulted the most potent dual inhibitor exhibiting IC50s of 0.53 and 2.90 µM against the RNase H and RDDP functions. Mutagenesis and docking studies suggested that compound 22 binds two allosteric pockets within the RT, one located between the RNase H active site and the primer grip region and the other close to the DNA polymerase catalytic centre.


Subject(s)
Anti-HIV Agents/pharmacology , HIV/drug effects , Oxazines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Thiophenes/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , HIV/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Oxazines/chemical synthesis , Oxazines/chemistry , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
15.
Chem Biol Drug Des ; 93(4): 582-589, 2019 04.
Article in English | MEDLINE | ID: mdl-30560566

ABSTRACT

Human immunodeficiency virus (HIV) reverse transcriptase (RT)-associated ribonuclease H (RNase H) remains as the only enzyme encoded within the viral genome not targeted by current antiviral drugs. In this work, we report the design, synthesis, and biologic evaluation of a novel series of galloyl derivatives with HIV-1 RNase H inhibitory activity. Most of them showed IC50 s at sub- to low-micromolar concentrations in enzymatic assays. The most potent compound was II-25 that showed an IC50 of 0.72 ± 0.07 µM in RNase H inhibition assays carried out with the HIV-1BH10 RT. II-25 was 2.8 times more potent than ß-thujaplicinol in these assays. Interestingly, II-25 and other galloyl derivatives were also found to inhibit the HIV IN strand transfer activity in vitro. Structure-activity relationships (SAR) studies and molecular modeling analysis predict key interactions with RT residues His539 and Arg557, while providing helpful insight for further optimization of selected compounds.


Subject(s)
Anti-HIV Agents/chemical synthesis , Drug Design , HIV-1/enzymology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Tropolone/analogs & derivatives , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Benzoic Acid/chemistry , Binding Sites , Catalytic Domain , HIV-1/drug effects , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship , Tropolone/chemical synthesis , Tropolone/chemistry , Tropolone/pharmacology
16.
Eur J Med Chem ; 156: 652-665, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30031976

ABSTRACT

Human immunodeficiency virus (HIV) reverse transcriptase (RT) associated ribonuclease H (RNase H) remains the only virally encoded enzymatic function not targeted by current drugs. Although a few chemotypes have been reported to inhibit HIV RNase H in biochemical assays, their general lack of significant antiviral activity in cell culture necessitates continued efforts in identifying highly potent RNase H inhibitors to confer antiviral activity. We report herein the design, synthesis, biochemical and antiviral evaluations of a new 6-arylthio subtype of the 3-hydroxypyrimidine-2,4-dione (HPD) chemotype. In biochemical assays these new analogues inhibited RT RNase H in single-digit nanomolar range without inhibiting RT polymerase (pol) at concentrations up to 10 µM, amounting to exceptional biochemical inhibitory selectivity. Many analogues also inhibited integrase strand transfer (INST) activity in low to sub micromolar range. More importantly, most analogues inhibited HIV in low micromolar range without cytotoxicity. In the end, compound 13j (RNase H IC50 = 0.005 µM; RT pol IC50 = 10 µM; INST IC50 = 4.0 µM; antiviral EC50 = 7.7 µM; CC50 > 100 µM) represents the best analogues within this series. These results characterize the new 6-arylthio-HPD subtype as a promising scaffold for HIV RNase H inhibitor discovery.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Cell Line , Drug Design , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/enzymology , Humans , Models, Molecular , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
17.
Eur J Med Chem ; 156: 680-691, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30031978

ABSTRACT

Human immunodeficiency virus (HIV) reverse transcriptase (RT)-associated ribonuclease H (RNase H) remains an unvalidated drug target. Reported HIV RNase H inhibitors generally lack significant antiviral activity. We report herein the design, synthesis, biochemical and antiviral evaluations of a new 6-biphenylmethyl subtype of the 3-hydroxypyrimidine-2,4-dione (HPD) chemotype. In biochemical assays, analogues of this new subtype potently inhibited RT RNase H in low nanomolar range without inhibiting RT polymerase (pol) or integrase strand transfer (INST) at the highest concentrations tested. In cell-based assays, a few analogues inhibited HIV in low micromolar range without cytotoxicity at concentrations up to 100 µM.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Catalytic Domain/drug effects , Cell Line , Drug Design , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/enzymology , Humans , Methylation , Models, Molecular , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Structure-Activity Relationship
18.
Phys Chem Chem Phys ; 20(37): 23873-23884, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-29947629

ABSTRACT

The rapid emergence of drug-resistant variants is one of the most common causes of highly active antiretroviral therapeutic (HAART) failure in patients infected with HIV-1. Compared with the existing HAART, the recently developed pyrrolyl diketo acid scaffold targeting both HIV-1 integrase (IN) and reverse transcriptase-associated ribonuclease H (RNase H) is an efficient approach to counteract the failure of anti-HIV treatment due to drug resistance. However, the binding mode and potential resistance profile of these inhibitors with important mechanistic principles remain poorly understood. To address this issue, an integrated computational method was employed to investigate the binding mode of inhibitor JMC6F with HIV-1 IN and RNase H. By using per-residue binding free energy decomposition analysis, the following residues: Asp64, Thr66, Leu68, Asp116, Tyr143, Gln148 and Glu152 in IN, Asp443, Glu478, Trp536, Lys541 and Asp549 in RNase H were identified as key residues for JMC6F binding. And then computational alanine scanning was carried to further verify the key residues. Moreover, the resistance profile of the currently known major mutations in HIV-1 IN and 2 mutations in RNase H against JMC6F was predicted by in silico mutagenesis studies. The results demonstrated that only three mutations in HIV-1 IN (Y143C, Q148R and N155H) and two mutations in HIV-1 RNase H (Y501R and Y501W) resulted in a reduction of JMC6F potency, thus indicating their potential role in providing resistance to JMC6F. These data provided important insights into the binding mode and resistance profile of the inhibitors with a pyrrolyl diketo acid scaffold in HIV-1 IN and RNase H, which would be helpful for the development of more effective dual HIV-1 IN and RNase H inhibitors.


Subject(s)
Anti-HIV Agents/chemistry , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Keto Acids/chemistry , Molecular Docking Simulation , Pyrroles/chemistry , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Integrase/genetics , HIV Integrase Inhibitors/pharmacology , Humans , Keto Acids/pharmacology , Mutation , Protein Binding , Pyrroles/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/genetics , Structure-Activity Relationship , Thermodynamics
19.
Curr Med Chem ; 25(14): 1682-1702, 2018.
Article in English | MEDLINE | ID: mdl-28088905

ABSTRACT

Combinations of antiretroviral drugs are successfully used to treat HIV-infected patients. However, drug resistance is a major problem that makes discovery of new antiretroviral drugs an ongoing priority. The ribonuclease H (RNase H) activity of the HIV-1 reverse transcriptase catalyzes the selective hydrolysis of the RNA strand of RNA:DNA heteroduplex replication intermediates, and represents an attractive unexploited target for drug development. This review reports on recent progress in the characterization of HIV-1 RNase H inhibitors from 2013 to 2016, describing their chemical structures, structureactivity relationship and binding modes. Focus is given to emerging medicinal chemistry principles and insights into the discovery and development of RNase H inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Anti-HIV Agents/chemistry , Cell Line, Tumor , Drug Design , HIV-1/metabolism , Humans , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship
20.
Viruses ; 9(11)2017 11 08.
Article in English | MEDLINE | ID: mdl-29117130

ABSTRACT

The South African national treatment programme includes nucleoside reverse transcriptase inhibitors (NRTIs) in both first and second line highly active antiretroviral therapy regimens. Mutations in the RNase H domain have been associated with resistance to NRTIs but primarily in HIV-1 subtype B studies. Here, we investigated the prevalence and association of RNase H mutations with NRTI resistance in sequences from HIV-1 subtype C infected individuals. RNase H sequences from 112 NRTI treated but virologically failing individuals and 28 antiretroviral therapy (ART)-naive individuals were generated and analysed. In addition, sequences from 359 subtype C ART-naive sequences were downloaded from Los Alamos database to give a total of 387 sequences from ART-naive individuals for the analysis. Fisher's exact test was used to identify mutations and Bayesian network learning was applied to identify novel NRTI resistance mutation pathways in RNase H domain. The mutations A435L, S468A, T470S, L484I, A508S, Q509L, L517I, Q524E and E529D were more prevalent in sequences from treatment-experienced compared to antiretroviral treatment naive individuals, however, only the E529D mutation remained significant after correction for multiple comparison. Our findings suggest a potential interaction between E529D and NRTI-treatment; however, site-directed mutagenesis is needed to understand the impact of this RNase H mutation.


Subject(s)
HIV-1/genetics , Mutation , Reverse Transcriptase Inhibitors/therapeutic use , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/genetics , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Bayes Theorem , Databases, Genetic , Drug Resistance, Viral/genetics , Genotype , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/drug effects , Humans , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...