Design, Synthesis, Biochemical, and Antiviral Evaluations of C6 Benzyl and C6 Biarylmethyl Substituted 2-Hydroxylisoquinoline-1,3-diones: Dual Inhibition against HIV Reverse Transcriptase-Associated RNase H and Polymerase with Antiviral ActivitiesClick to copy article linkArticle link copied!
- Sanjeev Kumar V. Vernekar
- Zheng Liu
- Eva Nagy
- Lena Miller
- Karen A. Kirby
- Daniel J. Wilson
- Jayakanth Kankanala
- Stefan G. Sarafianos
- Michael A. Parniak
- Zhengqiang Wang
Abstract
Reverse transcriptase (RT) associated ribonuclease H (RNase H) remains the only virally encoded enzymatic function not targeted by current chemotherapy against human immunodeficiency virus (HIV). Although numerous chemotypes have been reported to inhibit HIV RNase H biochemically, few show significant antiviral activity against HIV. We report herein the design, synthesis, and biological evaluations of a novel variant of 2-hydroxyisoquinoline-1,3-dione (HID) scaffold featuring a crucial C-6 benzyl or biarylmethyl moiety. The synthesis involved a recently reported metal-free direct benzylation between tosylhydrazone and boronic acid, which allowed the generation of structural diversity for the hydrophobic aromatic region. Biochemical studies showed that the C-6 benzyl and biarylmethyl HID analogues, previously unknown chemotypes, consistently inhibited HIV RT-associated RNase H and polymerase with IC50s in low to submicromolar range. The observed dual inhibitory activity remained uncompromised against RT mutants resistant to non-nucleoside RT inhibitors (NNRTIs), suggesting the involvement of binding site(s) other than the NNRTI binding pocket. Intriguingly, these same compounds inhibited the polymerase, but not the RNase H function of Moloney Murine Leukemia Virus (MoMLV) RT and also inhibited Escherichia coli RNase H. Additional biochemical testing revealed a substantially reduced level of inhibition against HIV integrase. Molecular docking corroborates favorable binding of these analogues to the active site of HIV RNase H. Finally, a number of these analogues also demonstrated antiviral activity at low micromolar concentrations.
Introduction
Figure 1
Figure 1. Major chemotypes reported as HIV RNase H active site inhibitors. Chemotypes 4–7 reflect a pharmacophore model consisting of a chelating triad (magenta) and an aryl or biaryl moiety (cyan) connected through a methylene or amino linker.
Figure 2
Figure 2. Design of a novel HID scaffold 9 based on the pharmacophore model of 4–7.
Results and Discussion
Chemistry
Scheme 1
Scheme aReagents and conditions: (a) NaBH4, THF, rt, 12 h, 87%; (b) n-BuLi, DMF, THF, −78 °C–rt, overnight, 76%; (c) TsNHNH2, toluene, 80 °C, 2 h; (d) boronic acid, K2CO3, 1,4-dioxane, 110 °C, 3–5 h, 40–55%; (e) PPTS, toluene, reflux, 6–12 h, 75–90%; (f) OsO4, NMO, t-BuOH/acetone/H2O, rt, 2–6 h, 60–81%; (g) NaIO4, RuCl3, CH3CN/CCl4/H2O, rt, 2–4 h, 67–85%; (h) ArB(OH)2, Pd(PPh3)4, K2CO3, EtOH/H2O, microwave, 120–150 °C, 20–30 min, 60–75%; (i) NH2OTHP, CDI, toluene, reflux, 12 h; (j) p-TSA hydrate, MeOH, 2–3 h, rt, 35–55% over two steps.
Biology
All New C6 Benzyl and C6 Biarylmethyl HID Analogues Potently Inhibit HIV RT Associated RNase H and Polymerase



IC50: concentration of a compound producing 50% inhibition, expressed as mean ± standard deviation from at least three independent experiments.
Substrate that measures internal cleavage.
Substrate that measures DNA 3′ end directed cleavage.
Substrate that measures RNA 5′ end directed cleavage.
Reconstituted HIV RNase H domain.


IC50: concentration of a compound producing 50% inhibition, expressed as mean ± standard deviation from at least three independent experiments.
RNase H substrates as described in the Experimental Section.


IC50: concentration of a compound producing 50% inhibition, expressed as mean ± standard deviation from two independent experiments of duplicates.
Some C6 Benzyl and C6 Biarylmethyl HID Analogues Moderately Inhibit HIV IN


Data with HTS-1 as substrate.
Expressed as mean ± standard deviation from at least three independent experiments.
Selectivity index defined by IC50 IN/IC50 RNase H.
Many C6 Benzyl and C6 Biarylmethyl HID Analogues Inhibit HIV-1 in Low Micromolar Range


All values are averages of two separate determinations.
50 μM was the highest concentration tested.
n.d., not determined.
RNase H Active Site Binding
Figure 3
Figure 3. Binding mode of compound 20i. Left: structure of full length RT with two subunits p66 and p51 (orange). p66 subunit comprises fingers (blue), palm (red), thumb (green), connection (yellow), and RNase H (magenta) domains. Compounds 7 (green) and 20i (cyan) are docked into the RNase H active site. Right: a close-up view of RNase H active site with predicted binding mode of compound 7 (green) and 20i (cyan). Metal cofactors (Mn2+) are colored in gray, with the active site residues (D443, E478, D498 and D549) in yellow. Pictures were generated using PyMol. (41)
Conclusions
Experimental Section
Chemistry: General Procedures
General Procedure 1 for the One-Pot Reductive Coupling of Aldehydes with Boronic Acids (14a–g)
5-Benzyl-2,3-dihydro-1H-inden-1-ol (14a)
General Procedure 2 for the Synthesis of Alkene (15a–g)
6-Benzyl-1H-indene (15a)
General Procedure 3 for the Synthesis of Diol (16a–g)
5-Benzyl-2,3-dihydro-1H-indene-1,2-diol (16a)
General Procedure 4 for the Synthesis of Dicarboxylic Acid (17a–g)
4-Benzyl-2-(carboxymethyl)benzoic Acid (17a)
General Procedure 5 for Suzuki Coupling (18h–t)
4-([1,1′-Biphenyl]-4-ylmethyl)-2-(carboxymethyl)benzoic Acid (18i)
General Procedure 6 for Cyclization and Deprotection (20a–t)
6-Benzyl-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20a)
2-Hydroxy-6-(4-methylbenzyl)isoquinoline-1,3(2H,4H)-dione (20b)
6-(4-Fluorobenzyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20c)
6-(4-Chlorobenzyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20d)
6-(4-Bromobenzyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20e)
2-Hydroxy-6-(4-(trifluoromethyl)benzyl)isoquinoline-1,3(2H,4H)-dione (20f)
6-(2,4-Difluorobenzyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20g)
6-(4-Cyclopropylbenzyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20h)
6-([1,1′-Biphenyl]-4-ylmethyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20i)
2-Hydroxy-6-(4-(thiophen-2-yl)benzyl)isoquinoline-1,3(2H,4H)-dione (20j)
6-((4′-Fluoro-[1,1′-biphenyl]-4-yl)methyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20k)
4′-((2-Hydroxy-1,3-dioxo-1,2,3,4-tetrahydroisoquinolin-6-yl)methyl)-[1,1′-biphenyl]-4-carbonitrile (20l)
2-Hydroxy-6-((4′-methoxy-[1,1′-biphenyl]-4-yl)methyl)isoquinoline-1,3(2H,4H)-dione (20m)
2-Hydroxy-6-((4′-(methylsulfonyl)-[1,1′-biphenyl]-4-yl)methyl)isoquinoline-1,3(2H,4H)-dione (20n)
4′-((2-Hydroxy-1,3-dioxo-1,2,3,4-tetrahydroisoquinolin-6-yl)methyl)-[1,1′-biphenyl]-4-sulfonamide (20o)
4′-((2-Hydroxy-1,3-dioxo-1,2,3,4-tetrahydroisoquinolin-6-yl)methyl)-[1,1′-biphenyl]-4-carboxamide (20p)
6-((2′,4′-Difluoro-[1,1′-biphenyl]-4-yl)methyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20q)
6-((2′,4′-Bis(trifluoromethyl)-[1,1′-biphenyl]-4-yl)methyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20r)
6-((3′-Fluoro-4′-methoxy-[1,1′-biphenyl]-4-yl)methyl)-2-hydroxyisoquinoline-1,3(2H,4H)-dione (20s)
4-(4-((2-Hydroxy-1,3-dioxo-1,2,3,4-tetrahydroisoquinolin-6-yl)methyl)phenyl)pyridin-1-ium 4-methylbenzenesulfonate (20t)
Biology
Reagents
Biologicals
Chemicals
RNase H Assay
RT Polymerase Assay
HIV IN Assay
Antiviral Assays
Modeling and Docking
Supporting Information
Characterization data, including 1H NMR, 13C NMR, and HRMS data, of all intermediates. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
This research was supported in part by the National Institutes of Health (AI100890 to S.G.S., M.A.P., and Z.W.), and by the Research Development and Seed Grant Program of the Center for Drug Design, University of Minnesota.
RT | reverse transcriptase |
HIV | human immunodeficiency virus |
RNase H | ribonuclease H |
HID | 2-hydroxyisoquinoline-1,3-dione |
IN | integrase |
NRTIs | nucleoside RT inhibitors |
NNRTIs | nonnucleoside RT inhibitors |
WT | wild-type |
DMF | N,N-dimethylformamide |
PPTS | pyridinium p-toluenesulfonate |
THP | tetrahydropyran |
p-TSA | p-toluenesulfonic acid |
SAR | structure–activity relationship |
RISF | retroviral integrase superfamily |
ST | strand transfer |
References
This article references 55 other publications.
- 1AIDSinfo; UNAIDS: Geneva 2013; http://www.unaids.org/en/dataanalysis/datatools/aidsinfo/.Google ScholarThere is no corresponding record for this reference.
- 2Barouch, D. H. Challenges in the development of an HIV-1 vaccine Nature 2008, 455, 613– 619Google ScholarThere is no corresponding record for this reference.
- 3Schiffner, T.; Sattentau, Q. J.; Dorrell, L. Development of prophylactic vaccines against HIV-1 Retrovirology 2013, 10, 72Google ScholarThere is no corresponding record for this reference.
- 4Zhang, J.; Crumpacker, C. Eradication of HIV and Cure of AIDS, Now and How? Front. Immunol. 2013, 4, 337Google Scholar4Eradication of HIV and Cure of AIDS, Now and How?Zhang Jielin; Crumpacker ClydeFrontiers in immunology (2013), 4 (), 337 ISSN:1664-3224.Recent studies have highlighted the importance of eradication of human immunodeficiency virus (HIV) and cure of acquired immunodeficiency syndrome (AIDS). However, a pivotal point that the patient immunity controls HIV reactivation after highly active anti-retroviral therapy [HAART or combination anti-retroviral therapy (cART)] remains less well addressed. In spite of the fact that both innate and adaptive immunities are indispensable and numerous cells participate in the anti-HIV immunity, memory CD4 T-cells are indisputably the key cells organizing all immune actions against HIV while being the targets of HIV. Here we present a view and multidisciplinary approaches to HIV/AIDS eradication and cure. We aim at memory CD4 T-cells, utilizing the stem cell properties of these cells to reprogram an anti-HIV memory repertoire to eliminate the viral reservoir, toward achieving an AIDS-free world.
- 5Siliciano, J. D.; Siliciano, R. F. HIV-1 eradication strategies: design and assessment Curr. Opin. HIV AIDS 2013, 8, 318– 325Google Scholar5HIV-1 eradication strategies: design and assessmentSiliciano Janet D; Siliciano Robert FCurrent opinion in HIV and AIDS (2013), 8 (4), 318-25 ISSN:.PURPOSE OF REVIEW: Recent developments have generated renewed interest in the possibility of curing HIV-1 infection. This review describes some of the practical challenges that will need to be overcome if curative strategies are to be successful. RECENT FINDINGS: The latent reservoir for HIV-1 in resting memory CD4 T cells is the major barrier to curing the infection. The most widely discussed approach to curing the infection involves finding agents that reverse latency in resting CD4 T cells, with the assumption that the cells will then die from viral cytopathic effects or be lysed by host cytolytic T lymphocytes (CTLs). A major challenge is the development of in-vitro models that can be used to explore mechanisms and identify latency-reversing agents (LRAs). Although several models have been developed, including primary cell models, none of them may fully capture the quiescent state of the cells that harbour latent HIV-1 in vivo. An additional problem is that LRAs that do not cause T-cell activation may not lead to the death of infected cells. Finally, measuring the effects of LRAs in vivo is complicated by the lack of correlation between different assays for the latent reservoir. SUMMARY: Progress on these practical issues is essential to finding a cure.
- 6Xing, S.; Siliciano, R. F. Targeting HIV latency: pharmacologic strategies toward eradication Drug Discovery Today 2013, 18, 541– 551Google ScholarThere is no corresponding record for this reference.
- 7Beilhartz, G. L.; Gotte, M. HIV-1 Ribonuclease H: Structure, Catalytic Mechanism and Inhibitors Viruses 2010, 2, 900– 926Google Scholar7HIV-1 ribonuclease H: structure, catalytic mechanism and inhibitorsBeilhartz, Greg L.; Gotte, MatthiasViruses (2010), 2 (), 900-926CODEN: VIRUBR; ISSN:1999-4915. (Molecular Diversity Preservation International)A review. Since the human immunodeficiency virus (HIV) was discovered as the etiol. agent of acquired immunodeficiency syndrome (AIDS), it has encouraged much research into antiviral compds. The reverse transcriptase (RT) of HIV has been a main target for antiviral drugs. However, all drugs developed so far inhibit the polymerase function of the enzyme, while none of the approved antiviral agents inhibit specifically the necessary RNase H (RNase H) function of RT. This review provides a background on structure-function relationships of HIV-1 RNase H, as well as an outline of current attempts to develop novel, potent chemotherapeutics against a difficult drug target.
- 8Sarafianos, S. G.; Marchand, B.; Das, K.; Himmel, D. M.; Parniak, M. A.; Hughes, S. H.; Arnold, E. Structure and function of HIV-1 reverse transcriptase: molecular mechanisms of polymerization and inhibition J. Mol. Biol. 2009, 385, 693– 713Google Scholar8Structure and function of HIV-1 reverse transcriptase: Molecular mechanisms of polymerization and inhibitionSarafianos, Stefan G.; Marchand, Bruno; Das, Kalyan; Himmel, Daniel M.; Parniak, Michael A.; Hughes, Stephen H.; Arnold, EddyJournal of Molecular Biology (2009), 385 (3), 693-713CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. The rapid replication of the HIV-1 virus and errors made during viral replication cause the virus to evolve rapidly in patients, making the problems of vaccine development and drug therapy particularly challenging. In the absence of an effective vaccine, drugs are the only useful treatment. Anti-HIV drugs work; so far drug therapy has saved more than 3 × 106 yr of life. Unfortunately, HIV-1 virus develops resistance to all of the available drugs. Although a no. of useful anti-HIV drugs have been approved for use in patients, the problems assocd. with drug toxicity and the development of resistance means that the search for new drugs is an ongoing process. The 3 viral enzymes, reverse transcriptase (RT), integrase (IN), and protease (PR) are all good drug targets. Two distinct types of RT inhibitors, both of which block the polymerase activity of RT, have been approved to treat HIV-1 infections, nucleoside analogs (NRTIs) and non-nucleosides (NNRTIs), and there are promising leads for compds. that either block the RNase H activity or block the polymerase in other ways. A better understanding of the structure and function(s) of RT and of the mechanism(s) of inhibition can be used to generate better drugs; in particular, drugs that are effective against the current drug-resistant strains of HIV-1.
- 9Cihlar, T.; Ray, A. S. Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudine Antiviral Res. 2010, 85, 39– 58Google Scholar9Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudineCihlar, Tomas; Ray, Adrian S.Antiviral Research (2010), 85 (1), 39-58CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. Twenty-five years ago, nucleoside analog 3'-azidothymidine (AZT) was shown to efficiently block the replication of HIV in cell culture. Subsequent studies demonstrated that AZT acts via the selective inhibition of HIV reverse transcriptase (RT) by its triphosphate metabolite. These discoveries have established the first class of antiretroviral agents: nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs). Over the years that followed, NRTIs evolved into the main component of antiretroviral drug combinations that are now used for the treatment of all populations of HIV infected patients. A total of thirteen NRTI drug products are now available for clin. application: eight individual NRTIs, four fixed-dose combinations of two or three NRTIs, and one complete fixed-dose regimen contg. two NRTIs and one non-nucleoside RT inhibitor. Multiple NRTIs or their prodrugs are in various stages of clin. development and new potent NRTIs are still being identified through drug discovery efforts. This article will review basic principles of the in vitro and in vivo pharmacol. of NRTIs, discuss their clin. use including limitations assocd. with long-term NRTI therapy, and describe newly identified NRTIs with promising pharmacol. profiles highlighting those in the development pipeline. This article forms part of a special issue of Antiviral Research marking the 25th anniversary of antiretroviral drug discovery and development, vol. 85, issue 1, 2010.
- 10de Bethune, M. P. Non-nucleoside reverse transcriptase inhibitors (NNRTIs), their discovery, development, and use in the treatment of HIV-1 infection: a review of the last 20 years (1989–2009) Antiviral Res. 2010, 85, 75– 90Google ScholarThere is no corresponding record for this reference.
- 11Corona, A.; Masaoka, T.; Tocco, G.; Tramontano, E.; Le Grice, S. F. Active site and allosteric inhibitors of the ribonuclease H activity of HIV reverse transcriptase Future Med. Chem. 2013, 5, 2127– 2139Google Scholar11Active site and allosteric inhibitors of the ribonuclease H activity of HIV reverse transcriptaseCorona, Angela; Masaoka, Takashi; Tocco, Graziella; Tramontano, Enzo; Le Grice, Stuart FJFuture Medicinal Chemistry (2013), 5 (18), 2127-2139CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. Despite the wealth of information available for the reverse transcriptase (RT)-assocd. RNase H (RNaseH) domain of lentiviruses, gamma-retroviruses and long terminal repeat contg. retrotransposons, exploiting this information in the form of an RNaseH inhibitor with high specificity and low cellular toxicity has been disappointing. However, it is now becoming increasingly evident that the two-subunit HIV-1 RT is a highly versatile enzyme, undergoing major structural alterations in order to interact with, position and ultimately hydrolyze the RNA component of an RNA/DNA hybrid. Thus, in addn. to targeting the RNaseH active site, identifying small mols. that bind elsewhere and disrupt catalysis allosterically by impairing conformational flexibility is gaining increased attention. This review summarizes current progress towards development of both active site and allosteric RNaseH inhibitors.
- 12Ilina, T.; Labarge, K.; Sarafianos, S. G.; Ishima, R.; Parniak, M. A. Inhibitors of HIV-1 Reverse Transcriptase-Associated Ribonuclease H Activity Biology (Basel, Switzerland) 2012, 1, 521– 541Google Scholar12Inhibitors of HIV-1 reverse transcriptase - associated ribonuclease H activityIlina, Tatiana; LaBarge, Krystal; Sarafianos, Stefan G.; Ishima, Rieko; Parniak, Michael A.Biology (Basel, Switzerland) (2012), 1 (3), 521-541CODEN: BBSIBX; ISSN:2079-7737. (MDPI AG)A review. HIV-1 enzyme reverse transcriptase (RT) is a major target for antiviral drug development, with over half of current FDA-approved therapeutics against HIV infection targeting the DNA polymerase activity of this enzyme. HIV-1 RT is a multifunctional enzyme that has RNA and DNA dependent polymerase activity, along with RNase H (RNase H) activity. The latter is responsible for degrdn. of the viral genomic RNA template during first strand DNA synthesis to allow completion of reverse transcription and the viral dsDNA. While the RNase H activity of RT has been shown to be essential for virus infectivity, all currently used drugs directed at RT inhibit the polymerase activity of the enzyme; none target RNase H. In the last decade, the increasing prevalence of HIV variants resistant to clin. used antiretrovirals has stimulated the search for inhibitors directed at stages of HIV replication different than those targeted by current drugs. HIV RNase H is one such novel target and, over the past few years, significant progress has been made in identifying and characterizing new RNase H inhibitor pharmacophores. In this review we focus mainly on the most potent low micromolar potency compds., as these provide logical bases for further development. We also discuss why HIV RNase H has been a difficult target for antiretroviral drug development.
- 13Klumpp, K.; Hang, J. Q.; Rajendran, S.; Yang, Y. L.; Derosier, A.; In, P. W. K.; Overton, H.; Parkes, K. E. B.; Cammack, N.; Martin, J. A. Two-metal ion mechanism of RNA cleavage by HIV RNase H and mechanism-based design of selective HIV RNase H inhibitors Nucleic Acids Res. 2003, 31, 6852– 6859Google Scholar13Two-metal ion mechanism of RNA cleavage by HIV RNase H and mechanism-based design of selective HIV RNase H inhibitorsKlumpp, Klaus; Hang, Julie Qi; Rajendran, Surendran; Yang, Yanli; Derosier, Andre; In, Philippe Wong Kai; Overton, Hilary; Parkes, Kevin E. B.; Cammack, Nick; Martin, Joseph A.Nucleic Acids Research (2003), 31 (23), 6852-6859CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Human immunodeficiency virus (HIV) RNase H activity is essential for the synthesis of viral DNA by HIV reverse transcriptase (HIV-RT). RNA cleavage by RNase H requires the presence of divalent metal ions, but the role of metal ions in the mechanism of RNA cleavage has not been resolved. We measured HIV RNase H activity assocd. with HIV-RT protein in the presence of different concns. of either Mg2+, Mn2+, Co2+ or a combination of these divalent metal ions. Polymerase-independent HIV RNase H was similar to or more active with Mn2+ and Co2+ compared with Mg2+. Activation of RNase H by these metal ions followed sigmoidal dose-response curves suggesting cooperative metal ion binding. Titrn. of Mg2+-bound HIV RNase H with Mn2+ or Co2+ ions generated bell-shaped activity dose-response curves. Higher activity could be achieved through simultaneous binding of more than one divalent metal ion at intermediate Mn2+ and Co2+ concns., and complete replacement of Mg2+ occurred at higher Mn2+ or Co2+ concns. These results are consistent with a two-metal ion mechanism of RNA cleavage as previously suggested for a no. of polymerase-assocd. nucleases. In contrast, the structurally highly homologous RNase HI from Escherichia coli is most strongly activated by Mg2+, is significantly inhibited by submillimolar concns. of Mn2+ and most probably cleaves RNA via a one-metal ion mechanism. Based on this difference in active site structure, a series of small mol. N-hydroxyimides was identified with significant enzyme inhibitory potency and selectivity for HIV RNase H.
- 14Himmel, D. M.; Sarafianos, S. G.; Dharmasena, S.; Hossain, M. M.; McCoy-Simandle, K.; Ilina, T.; Clark, A. D.; Knight, J. L.; Julias, J. G.; Clark, P. K.; Krogh-Jespersen, K.; Levy, R. M.; Hughes, S. H.; Parniak, M. A.; Arnold, E. HIV-1 reverse transcriptase structure with RNase H inhibitor dihydroxy benzoyl naphthyl hydrazone bound at a novel site ACS Chem. Biol. 2006, 1, 702– 712Google Scholar14HIV-1 Reverse Transcriptase Structure with RNase H Inhibitor Dihydroxy Benzoyl Naphthyl Hydrazone Bound at a Novel SiteHimmel, Daniel M.; Sarafianos, Stefan G.; Dharmasena, Sanjeewa; Hossain, Mohammed M.; McCoy-Simandle, Kessler; Ilina, Tatiana; Clark, Arthur D., Jr.; Knight, Jennifer L.; Julias, John G.; Clark, Patrick K.; Krogh-Jespersen, Karsten; Levy, Ronald M.; Hughes, Stephen H.; Parniak, Michael A.; Arnold, EddyACS Chemical Biology (2006), 1 (11), 702-712CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The rapid emergence of drug-resistant variants of human immunodeficiency virus, type 1 (HIV-1), has limited the efficacy of anti-acquired immune deficiency syndrome (AIDS) treatments, and new lead compds. that target novel binding sites are needed. The authors have detd. the 3.15 Å resoln. crystal structure of HIV-1 reverse transcriptase (RT) complexed with dihydroxy benzoyl naphthyl hydrazone (DHBNH), an HIV-1 RT RNase H (RNH) inhibitor (RNHI). DHBNH is effective against a variety of drug-resistant HIV-1 RT mutants. While DHBNH has little effect on most aspects of RT-catalyzed DNA synthesis, at relatively high concns. it does inhibit the initiation of RNA-primed DNA synthesis. Although primarily an RNHI, DHBNH binds >50 Å away from the RNH active site, at a novel site near both the polymerase active site and the nonnucleoside RT inhibitor (NNRTI) binding pocket. When DHBNH binds, both Tyr181 and Tyr188 remain in the conformations seen in unliganded HIV-1 RT. DHBNH interacts with conserved residues (Asp186, Trp229) and has substantial interactions with the backbones of several less well-conserved residues. On the basis of this structure, the authors designed substituted DHBNH derivs. that interact with the NNRTI-binding pocket. These compds. inhibit both the polymerase and RNH activities of RT.
- 15Fuji, H.; Urano, E.; Futahashi, Y.; Hamatake, M.; Tatsumi, J.; Hoshino, T.; Morikawa, Y.; Yamamoto, N.; Komano, J. Derivatives of 5-nitro-furan-2-carboxylic acid carbamoylmethyl ester inhibit RNase H activity associated with HIV-1 reverse transcriptase J. Med. Chem. 2009, 52, 1380– 1387Google ScholarThere is no corresponding record for this reference.
- 16Tramontano, E.; Esposito, F.; Badas, R.; Di Santo, R.; Costi, R.; La Colla, P. 6-[1-(4-Fluorophenyl)methyl-1H-pyrrol-2-yl)]-2,4-dioxo-5-hexenoic acid ethyl ester a novel diketo acid derivative which selectively inhibits the HIV-1 viral replication in cell culture and the ribonuclease H activity in vitro Antiviral Res. 2005, 65, 117– 124Google Scholar166-[1-(4-Fluorophenyl)methyl-1H-pyrrol-2-yl]-2,4-dioxo-5-hexenoic acid ethyl ester a novel diketo acid derivative which selectively inhibits the HIV-1 viral replication in cell culture and the ribonuclease H activity in vitroTramontano, Enzo; Esposito, Francesca; Badas, Roberta; Di Santo, Roberto; Costi, Roberta; La Colla, PaoloAntiviral Research (2005), 65 (2), 117-124CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)The human immunodeficiency virus-type 1 (HIV-1) reverse transcriptase (RT) is a multifunctional enzyme which displays DNA polymerase activity, which recognizes RNA and DNA templates, and a degradative RNase H activity. While both RT functions are required for retroviral replication, until now only the polymerase function has been widely explored as drug target. We have identified a novel diketo acid deriv., 6-[1-(4-fluorophenyl)methyl-1H-pyrrol-2-yl]-2,4-dioxo-5-hexenoic acid Et ester (RDS 1643), which inhibits in enzyme assays the HIV-1 RT-assocd. polymerase-independent RNase H activity but has no effect on the HIV-1 RT-assocd. RNA-dependent DNA polymerase (RDDP) activity and on the RNase H activities displayed by the Avian Myeloblastosis Virus and E. coli. Time-dependence studies revealed that the compd. is active independently on the order of its addn. to the reaction mixt., and inhibition kinetics studies demonstrated that RDS 1643 inhibits the RNase H activity noncompetitively, with a KI value of 17 μM. When RDS 1643 was combined with non-nucleoside RT inhibitors (NNRTI), such as efavirenz and nevirapine, results indicated that RDS 1643 does not affect the NNRTIs anti-RDDP activity and that, vice versa, the NNRTIs do not alter the RNase H inhibition by RDS 1643. When assayed on the viral replication in cell-based assays, RDS 1643 inhibited the HIV-1IIIB strain with an EC50 of 14 μM. Similar results were obtained against the Y181C and Y181C/K103N HIV-1 NNRTI resistant mutant strains. RDS 1643 may be the first HIV-1 inhibitor selectively targeted to the viral RT-assocd. RNase-H function.
- 17Lansdon, E. B.; Liu, Q.; Leavitt, S. A.; Balakrishnan, M.; Perry, J. K.; Lancaster-Moyer, C.; Kutty, N.; Liu, X.; Squires, N. H.; Watkins, W. J.; Kirschberg, T. A. Structural and binding analysis of pyrimidinol carboxylic acid and N-hydroxy quinazolinedione HIV-1 RNase H inhibitors Antimicrob. Agents Chemother. 2011, 55, 2905– 2915Google ScholarThere is no corresponding record for this reference.
- 18Williams, P. D.; Staas, D. D.; Venkatraman, S.; Loughran, H. M.; Ruzek, R. D.; Booth, T. M.; Lyle, T. A.; Wai, J. S.; Vacca, J. P.; Feuston, B. P.; Ecto, L. T.; Flynn, J. A.; DiStefano, D. J.; Hazuda, D. J.; Bahnck, C. M.; Himmelberger, A. L.; Dornadula, G.; Hrin, R. C.; Stillmock, K. A.; Witmer, M. V.; Miller, M. D.; Grobler, J. A. Potent and selective HIV-1 ribonuclease H inhibitors based on a 1-hydroxy-1,8-naphthyridin-2(1H)-one scaffold Bioorg. Med. Chem. Lett. 2010, 20, 6754– 6757Google ScholarThere is no corresponding record for this reference.
- 19Gerondelis, P.; Johns, B. A., The development of novel pyrido-pyrimidinone antiretrovirals with selective activity against HIV ribonuclease H. , Cold Spring Harbor Laboratories Conference on Retroviruses, Cold Spring Harbor, NY, May 21–26, 2012; Cold Spring Harbor Press: Cold Spring Harbor, NY, 2012.Google ScholarThere is no corresponding record for this reference.
- 20Beilhartz, G. L.; Ngure, M.; Johns, B. A.; DeAnda, F.; Gerondelis, P.; Gotte, M. Inhibition of the Ribonuclease H Activity of HIV-1 Reverse Transcriptase by GSK5750 Correlates with Slow Enzyme–Inhibitor Dissociation J. Biol. Chem. 2014, 289, 16270– 16277Google ScholarThere is no corresponding record for this reference.
- 21Chen, Y. L.; Tang, J.; Kesler, M. J.; Sham, Y. Y.; Vince, R.; Geraghty, R. J.; Wang, Z. Q. The design, synthesis and biological evaluations of C-6 or C-7 substituted 2-hydroxyisoquinoline-1,3-diones as inhibitors of hepatitis C virus Bioorg. Med. Chem. 2012, 20, 467– 479Google ScholarThere is no corresponding record for this reference.
- 22Billamboz, M.; Bailly, F.; Lion, C.; Calmels, C.; Andreola, M. L.; Witvrouw, M.; Christ, F.; Debyser, Z.; De Luca, L.; Chimirri, A.; Cotelle, P. 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as inhibitors of HIV-1 integrase and reverse transcriptase RNase H domain: influence of the alkylation of position 4 Eur. J. Med. Chem. 2011, 46, 535– 546Google ScholarThere is no corresponding record for this reference.
- 23Billamboz, M.; Suchaud, V.; Bailly, F.; Lion, C.; Demeulemeester, J.; Calmels, C.; Andreola, M. L.; Christ, F.; Debyser, Z.; Cotelle, P. 4-Substituted 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as a Novel Class of HIV-1 Integrase Inhibitors ACS Med. Chem. Lett. 2013, 4, 41– 46Google ScholarThere is no corresponding record for this reference.
- 24Desimmie, B. A.; Demeulemeester, J.; Suchaud, V.; Taltynov, O.; Billamboz, M.; Lion, C.; Bailly, F.; Strelkov, S. V.; Debyser, Z.; Cotelle, P.; Christ, F. 2-Hydroxyisoquinoline-1,3(2H,4H)-diones (HIDs), Novel Inhibitors of HIV Integrase with a High Barrier to Resistance ACS Chem. Biol. 2013, 8, 1187– 1194Google ScholarThere is no corresponding record for this reference.
- 25Suchaud, V.; Bailly, F.; Lion, C.; Calmels, C.; Andréola, M.-L.; Christ, F.; Debyser, Z.; Cotelle, P. Investigation of a Novel Series of 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as Human Immunodeficiency Virus Type 1 Integrase Inhibitors J. Med. Chem. 2014, 57, 4640– 4660http://dx.doi.org/10.1021/jm500109zGoogle ScholarThere is no corresponding record for this reference.
- 26Billamboz, M.; Bailly, F.; Lion, C.; Touati, N.; Vezin, H.; Calmels, C.; Andreola, M. L.; Christ, F.; Debyser, Z.; Cotelle, P. Magnesium Chelating 2-Hydroxyisoquinoline-1,3(2H,4H)-diones, as Inhibitors of HIV-1 Integrase and/or the HIV-1 Reverse Transcriptase Ribonuclease H Domain: Discovery of a Novel Selective Inhibitor of the Ribonuclease H Function J. Med. Chem. 2011, 54, 1812– 1824Google Scholar26Magnesium Chelating 2-Hydroxyisoquinoline-1,3(2H,4H)-diones, as Inhibitors of HIV-1 Integrase and/or the HIV-1 Reverse Transcriptase Ribonuclease H Domain: Discovery of a Novel Selective Inhibitor of the Ribonuclease H FunctionBillamboz, Muriel; Bailly, Fabrice; Lion, Cedric; Touati, Nadia; Vezin, Herve; Calmels, Christina; Andreola, Marie-Line; Christ, Frauke; Debyser, Zeger; Cotelle, PhilippeJournal of Medicinal Chemistry (2011), 54 (6), 1812-1824CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)2-Hydroxyisoquinoline-1,3(2H,4H)-dione was recently discovered as a scaffold for the inhibition of HIV-1 integrase and the RNase H function of HIV-1 reverse transcriptase. First, we investigate its interaction with Mg2+ and Mn2+ using different spectroscopic techniques and report that 2-hydroxyisoquinoline-1,3(2H,4H)-dione forms a 1:1 complex with Mg2+ but a 1:2 complex with Mn2+. The complex formation requires enolization of the ligand. ESR spectroscopy shows a redox reaction between the ligand and Mn2+ producing superoxide anions. Second, 2-hydroxyisoquinoline-1,3(2H,4H)-dione, its magnesium complex, and its 4-Me and 2-hydroxy-4-methoxycarbonylisoquinoline-1,3(2H,4H)-diones were tested as inhibitors of HIV-1 integrase, reverse transcriptase RNase H, and DNA polymerase functions. Their antiviral activities were evaluated and 2-hydroxy-4-methoxycarbonyl-isoquinoline-1,3(2H,4H)-dione was found to inhibit the viral replication of HIV-1 in MT-4 cells. Cross-resistance was measured for this compd. on three different viral strains. Exptl. data suggest that the antiviral activity of 2-hydroxy-4-methoxycarbonylisoquinoline-1,3(2H,4H)-dione is probably due to the RNase H inhibition.
- 27Billamboz, M.; Bailly, F.; Barreca, M. L.; De Luca, L.; Mouscadet, J. F.; Calmels, C.; Andreola, M. L.; Witvrouw, M.; Christ, F.; Debyser, Z.; Cotelle, P. Design, Synthesis, and Biological Evaluation of a Series of 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as Dual Inhibitors of Human Immunodeficiency Virus Type 1 Integrase and the Reverse Transcriptase RNase H Domain J. Med. Chem. 2008, 51, 7717– 7730Google ScholarThere is no corresponding record for this reference.
- 28Barluenga, J.; Tomas-Gamasa, M.; Aznar, F.; Valdes, C. Metal-free carbon–carbon bond-forming reductive coupling between boronic acids and tosylhydrazones Nature Chem. 2009, 1, 494– 499Google Scholar28Metal-free carbon-carbon bond-forming reductive coupling between boronic acids and tosylhydrazonesBarluenga, Jose; Tomas-Gamasa, Maria; Aznar, Fernando; Valdes, CarlosNature Chemistry (2009), 1 (6), 494-499, S494/1-S494/46CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The formation of carbon-carbon bonds is a fundamental transformation in org. synthesis. In spite of the myriad methods available, advantageous methodologies in terms of selectivity, availability of starting materials, operational simplicity, functional-group tolerance, environmental sustainability and economy are in const. demand. In this context, the development of new cross-coupling reactions that use catalysts based on inexpensive and non-toxic metals is attracting increasing attention. Similarly, efficient processes that do not require a metal catalyst are of extraordinary interest. Here, we report a new and efficient metal-free carbon-carbon bond-forming coupling between tosylhydrazones and boronic acids. This reaction is very general and functional-group tolerant. As the required tosylhydrazones are easily generated from carbonyl compds., it can be seen as a reductive coupling of carbonyls, a process of high synthetic relevance that requires several steps using other methodologies.
- 29Vanrheenen, V.; Cha, D. Y.; Hartley, W. M. Catalytic Osmium-Tetroxide Oxidation of Olefins—cis-1,2-Cyclohexanediol Org. Synth. 1988, 50–9, 342– 348Google ScholarThere is no corresponding record for this reference.
- 30Vanrheenen, V.; Kelly, R. C.; Cha, D. Y. Improved Catalytic OsO4 Oxidation of Olefins to cis-1,2-Glycols Using Tertiary Amine Oxides as Oxidant Tetrahedron Lett. 1976, 1973– 1976Google ScholarThere is no corresponding record for this reference.
- 31Yang, D.; Zhang, C. Ruthenium-catalyzed oxidative cleavage of olefins to aldehydes J. Org. Chem. 2001, 66, 4814– 4818Google Scholar31Ruthenium-Catalyzed Oxidative Cleavage of Olefins to AldehydesYang, Dan; Zhang, ChiJournal of Organic Chemistry (2001), 66 (14), 4814-4818CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Three oxidn. protocols have been developed to cleave olefins to carbonyl compds. with ruthenium trichloride as catalyst (3.5 mol %). These methods convert olefins that are not fully substituted to aldehydes rather than carboxylic acids. While aryl olefins were cleaved to arom. aldehydes in excellent yields by using the system of RuCl3-Oxone-NaHCO3 in CH3CN-H2O (1.5:1), aliph. olefins were converted into alkyl aldehydes with RuCl3-NaIO4 in 1,2-dichloroethane-H2O (1:1) in good to excellent yields. It is noteworthy that terminal aliph. olefins were cleaved to the corresponding aldehydes in excellent yields by using RuCl3-NaIO4 in CH3CN-H2O (6:1).
- 32Parniak, M. A.; Min, K. L.; Budihas, S. R.; Le Grice, S. F.; Beutler, J. A. A fluorescence-based high-throughput screening assay for inhibitors of human immunodeficiency virus-1 reverse transcriptase-associated ribonuclease H activity Anal. Biochem. 2003, 322, 33– 39Google ScholarThere is no corresponding record for this reference.
- 33Parniak, M. A.; Min, K. L. Substrate for assaying ribonuclease H activity. US 7,186,520 March6, 2007.Google ScholarThere is no corresponding record for this reference.
- 34Parniak, M. A.; Min, K. L. Method of identifying or characterizing a compound that modulates ribonuclease H activity. US 7,439,035 October21, 2008.Google ScholarThere is no corresponding record for this reference.
- 35Nowotny, M. Retroviral integrase superfamily: the structural perspective EMBO Rep. 2009, 10, 144– 151Google Scholar35Retroviral integrase superfamily: the structural perspectiveNowotny, MarcinEMBO Reports (2009), 10 (2), 144-151CODEN: ERMEAX; ISSN:1469-221X. (Nature Publishing Group)A review. The retroviral integrase superfamily (RISF) comprises numerous important nucleic acid-processing enzymes, including transposases, integrases and various nucleases. These enzymes are involved in a wide range of processes such as transposition, replication and repair of DNA, homologous recombination, and RNA-mediated gene silencing. Two out of the four enzymes that are encoded by the human immunodeficiency virus, RNase H1 and integrase, are members of this superfamily. RISF enzymes act on various substrates, and yet show remarkable mechanistic and structural similarities. All share a common fold of the catalytic core and the active site, which is composed primarily of carboxylate residues. Here, RISF proteins are presented from a structural perspective, describing the individual members and the common and divergent elements of their structures, as well as the mechanistic insights gained from the structures of RNase H1 enzyme complexes with RNA/DNA hybrids.
- 36Barreca, M. L.; Ferro, S.; Rao, A.; De Luca, L.; Zappala, M.; Monforte, A. M.; Debyser, Z.; Witvrouw, M.; Chimirri, A. Pharmacophore-based design of HIV-1 integrase strand-transfer inhibitors J. Med. Chem. 2005, 48, 7084– 7088Google ScholarThere is no corresponding record for this reference.
- 37Abram, M. E.; Parniak, M. A. Virion instability of human immunodeficiency virus type 1 reverse transcriptase (RT) mutated in the protease cleavage site between RT p51 and the RT RNase H domain J. Virol. 2005, 79, 11952– 11961Google Scholar37Virion instability of human immunodeficiency virus type 1 reverse transcriptase (RT) mutated in the protease cleavage site between RT p51 and the RT RNase H domainAbram, Michael E.; Parniak, Michael A.Journal of Virology (2005), 79 (18), 11952-11961CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Each of the human immunodeficiency virus type 1 (HIV-1) pol-encoded enzymes, protease (PR), reverse transcriptase (RT), and integrase (IN), is active only as a dimer (or higher-order oligomer in the case of IN), but only RT comprises subunits of different masses. RT is a heterodimer of 66-kDa and 51-kDa subunits. The latter is formed by HIV PR-catalyzed cleavage of p66 during virion maturation, resulting in the removal of the RNase H (RNH) domain of a p66 subunit. In order to study the apparent need for RT heterodimers in the context of the virion, we introduced a variety of mutations in the RT p51-RNH protease cleavage site of an infectious HIV-1 mol. clone. Surprisingly, rather than leading to virions with increased RT p66 content, most of the mutations resulted in significantly attenuated virus that contained greatly decreased levels of RT that in many cases was primarily p51 RT. IN levels were also reduced in several mutants. However, most mutants showed normal levels of the Pr160gag-pol precursor polyprotein, suggesting that reduced virion RT arose from proteolytic instability rather than decreased incorporation. Mutant virion p24 Gag levels were equiv. to wild type, indicating that Gag incorporation and processing were not affected. Repeated passage of MT-2 cells exposed to mutant viruses led to the appearance of virus with improved replication capacity; these virions contained normally processed RT at near-wild-type levels. These results imply that addnl. proteolytic processing of RT to the p66/p51 heterodimer is essential to provide proteolytic stability of RT during HIV-1 maturation.
- 38Parikh, U. M.; Koontz, D. L.; Chu, C. K.; Schinazi, R. F.; Mellors, J. W. In vitro activity of structurally diverse nucleoside analogs against human immunodeficiency virus type 1 with the K65R mutation in reverse transcriptase Antimicrob. Agents Chemother. 2005, 49, 1139– 1144Google ScholarThere is no corresponding record for this reference.
- 39Su, H.-P.; Yan, Y.; Prasad, G. S.; Smith, R. F.; Daniels, C. L.; Abeywickrema, P. D.; Reid, J. C.; Loughran, H. M.; Kornienko, M.; Sharma, S.; Grobler, J. A.; Xu, B.; Sardana, V.; Allison, T. J.; Williams, P. D.; Darke, P. L.; Hazuda, D. J.; Munshi, S. Structural Basis for the Inhibition of RNase H Activity of HIV-1 Reverse Transcriptase by RNase H Active Site-Directed Inhibitors J. Virol. 2010, 84, 7625– 7633Google Scholar39Structural basis for the inhibition of RNase H activity of HIV-1 reverse transcriptase by RNase H active site-directed inhibitorsSu, Hua-Poo; Yan, Youwei; Prasad, G. Sridhar; Smith, Robert F.; Daniels, Christopher L.; Abeywickrema, Pravien D.; Reid, John C.; Loughran, H. Marie; Kornienko, Maria; Sharma, Sujata; Grobler, Jay A.; Xu, Bei; Sardana, Vinod; Allison, Timothy J.; Williams, Peter D.; Darke, Paul L.; Hazuda, Daria J.; Munshi, SanjeevJournal of Virology (2010), 84 (15), 7625-7633CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)HIV/AIDS continues to be a menace to public health. Several drugs currently on the market have successfully improved the ability to manage the viral burden in infected patients. However, new drugs are needed to combat the rapid emergence of mutated forms of the virus that are resistant to existing therapies. Currently, approved drugs target three of the four major enzyme activities encoded by the virus that are crit. to the HIV life cycle. Although a no. of inhibitors of HIV RNase H activity have been reported, few inhibit by directly engaging the RNase H active site. Here, we describe structures of naphthyridinone-contg. inhibitors bound to the RNase H active site. This class of compds. binds to the active site via two metal ions that are coordinated by catalytic site residues, D443, E478, D498, and D549. The directionality of the naphthyridinone pharmacophore is restricted by the ordering of D549 and H539 in the RNase H domain. In addn., one of the naphthyridinone-based compds. was found to bind at a second site close to the polymerase active site and non-nucleoside/nucleotide inhibitor sites in a metal-independent manner. Further characterization, using fluorescence-based thermal denaturation and a crystal structure of the isolated RNase H domain reveals that this compd. can also bind the RNase H site and retains the metal-dependent binding mode of this class of mols. These structures provide a means for structurally guided design of novel RNase H inhibitors.
- 40Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes J. Med. Chem. 2006, 49, 6177– 6196Google Scholar40Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 41PyMOL, The PyMOL Molecular Graphics System, version 1.5.0.4; Schrödinger, LLC: New York 2013.Google ScholarThere is no corresponding record for this reference.
- 42Fletcher, R. S.; Holleschak, G.; Nagy, E.; Arion, D.; Borkow, G.; Gu, Z.; Wainberg, M. A.; Parniak, M. A. Single-step purification of recombinant wild-type and mutant HIV-1 reverse transcriptase Protein Express. Purif. 1996, 7, 27– 32Google Scholar42Single-step purification of recombinant wild-type and mutant HIV-1 reverse transcriptaseFletcher, Ronald S.; Holleschak, Gyula; Nagy, Eva; Arion, Dominique; Borkow, Gadi; Gu, Zhengxian; Wainberg, Mark A.; Parniak, Michael A.Protein Expression and Purification (1996), 7 (1), 27-32CODEN: PEXPEJ; ISSN:1046-5928. (Academic)A single-step method that enables the purifn. of HIV-1 recombinant reverse transcriptase (RT) directly from bacterial lysates in <2 h is described. Clarified lysates were applied to com. Q- and S-matrix cartridge columns connected in series. The columns were washed with low-salt buffer to remove unbound protein, then the Q column was removed and RT was eluted from the S column using a salt gradient. The purifn. was carried out with both medium-pressure and high-pressure chromatog. systems. The purifns. were carried out at room temp. near neutral pH, providing enzyme with high DNA polymerase specific activity. A crucial aspect of the procedure was the use of Tris buffer, a buffer that is normally incompatible in cation-exchange methods. The method was applicable for the purifn. of the p51/p66 heterodimer and the p51 and p66 homodimer forms of RT. This method was used to purify wild-type RT and several recombinant RT proteins contg. mutations correlated with dideoxynucleoside drug resistance.
- 43Gong, Q.; Menon, L.; Ilina, T.; Miller, L. G.; Ahn, J.; Parniak, M. A.; Ishima, R. Interaction of HIV-1 reverse transcriptase ribonuclease H with an acylhydrazone inhibitor Chem. Biol. Drug Des. 2011, 77, 39– 47Google Scholar43Interaction of HIV-1 reverse transcriptase ribonuclease H with an acylhydrazone inhibitorGong, Qingguo; Menon, Lakshmi; Ilina, Tatiana; Miller, Lena G.; Ahn, Jinwoo; Parniak, Michael A.; Ishima, RiekoChemical Biology & Drug Design (2010), 77 (1), 39-47CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)HIV-1 reverse transcriptase is a bifunctional enzyme, having both DNA polymerase (RNA- and DNA-dependent) and RNase H activities. HIV-1 reverse transcriptase has been an exceptionally important target for antiretroviral therapeutic development, and nearly half of the current clin. used antiretrovirals target reverse transcriptase DNA polymerase. However, no inhibitors of reverse transcriptase RNase H are on the market or in preclin. development. Several drug-like small mol. inhibitors of reverse transcriptase RNase H have been described, but little structural information is available about the interactions between reverse transcriptase RNase H and inhibitors that exhibit antiviral activity. In this report, we describe NMR studies of the interaction of a new RNase H inhibitor, BHMP07, with a catalytically active HIV-1 reverse transcriptase RNase H domain fragment. We carried out soln. NMR expts. to identify the interaction interface of BHMP07 with the RNase H domain fragment. Chem. shift changes of backbone amide signals at different BHMP07 concns. clearly demonstrate that BHMP07 mainly recognizes the substrate handle region in the RNase H fragment. Using RNase H inhibition assays and reverse transcriptase mutants, the binding specificity of BHMP07 was compared with another inhibitor, dihydroxy benzoyl naphthyl hydrazone. Our results provide a structural characterization of the RNase H inhibitor interaction and are likely to be useful for further improvements of the inhibitors.
- 44Dabora, J. M.; Marqusee, S. Equilibrium unfolding of Escherichia coli ribonuclease H: characterization of a partially folded state Protein Sci. 1994, 3, 1401– 1408Google Scholar44Equilibrium unfolding of Escherichia coli ribonuclease H: characterization of a partially folded stateDabora, Jonathan M.; Marqusee, SusanProtein Science (1994), 3 (9), 1401-8CODEN: PRCIEI; ISSN:0961-8368.The equil. unfolding of Escherichia coli RNase HI (RNase H), a member of a family of enzymes the cleaves RNA from RNA:DNA hybrids, was examd. A completely synthetic gene was constructed that expresses a variant of the wild-type sequence with all 3 cysteines replaced with alanine. The resulting recombinant protein is active and folds reversibly. Denaturation studies monitored by CD and tryptophan fluorescence yield coincident curves that suggest that the equil. unfolding reaction is a 2-state process. Acid denaturation, however, reveals a cooperative transition at pH ∼1.8 to a partially folded state. This acid state can be further denatured in a reversible manner by the addn. of heat or urea as monitored by either CD or tryptophan fluorescence. Anal. ultracentrifugation studies indicate that the acid state of RNase H is both compact and monomeric. Although compact, the acid state does not resemble the native protein: the acid state displays a near-UV CD spectrum similar to the unfolded state and binds to and enhances the fluorescence of the dye 1-anilinonaphthalene 8-sulfonate much more than either the native or unfolded states. Therefore, the acid state of E. coli RNase H has the characteristics of a molten globule: it retains a high degree of secondary structure, remains compact, yet does not appear to contain a tightly packed core.
- 45Ndongwe, T. P.; Adedeji, A. O.; Michailidis, E.; Ong, Y. T.; Hachiya, A.; Marchand, B.; Ryan, E. M.; Rai, D. K.; Kirby, K. A.; Whatley, A. S.; Burke, D. H.; Johnson, M.; Ding, S.; Zheng, Y. M.; Liu, S. L.; Kodama, E.; Delviks-Frankenberry, K. A.; Pathak, V. K.; Mitsuya, H.; Parniak, M. A.; Singh, K.; Sarafianos, S. G. Biochemical, inhibition and inhibitor resistance studies of xenotropic murine leukemia virus-related virus reverse transcriptase Nucleic Acids Res. 2012, 40, 345– 359Google ScholarThere is no corresponding record for this reference.
- 46Kirby, K. A.; Marchand, B.; Ong, Y. T.; Ndongwe, T. P.; Hachiya, A.; Michailidis, E.; Leslie, M. D.; Sietsema, D. V.; Fetterly, T. L.; Dorst, C. A.; Singh, K.; Wang, Z.; Parniak, M. A.; Sarafianos, S. G. Structural and inhibition studies of the RNase H function of xenotropic murine leukemia virus-related virus reverse transcriptase Antimicrob. Agents Chemother. 2012, 56, 2048– 2061Google ScholarThere is no corresponding record for this reference.
- 47Michailidis, E.; Marchand, B.; Kodama, E. N.; Singh, K.; Matsuoka, M.; Kirby, K. A.; Ryan, E. M.; Sawani, A. M.; Nagy, E.; Ashida, N.; Mitsuya, H.; Parniak, M. A.; Sarafianos, S. G. Mechanism of inhibition of HIV-1 reverse transcriptase by 4′-ethynyl-2-fluoro-2′-deoxyadenosine triphosphate, a translocation-defective reverse transcriptase inhibitor J. Biol. Chem. 2009, 284, 35681– 35691Google ScholarThere is no corresponding record for this reference.
- 48Wang, Z.; Bennett, E. M.; Wilson, D. J.; Salomon, C.; Vince, R. Rationally designed dual inhibitors of HIV reverse transcriptase and integrase J. Med. Chem. 2007, 50, 3416– 3419Google ScholarThere is no corresponding record for this reference.
- 49Sirivolu, V. R.; Vernekar, S. K. V.; Ilina, T.; Myshakina, N. S.; Parniak, M. A.; Wang, Z. Q. Clicking 3′-Azidothymidine into Novel Potent Inhibitors of Human Immunodeficiency Virus J. Med. Chem. 2013, 56, 8765– 8780Google ScholarThere is no corresponding record for this reference.
- 50HIV-1 reverse transcriptase with inhibitor; RCSB Protein Data Bank: Piscataway, NJ, 2010; http://www.rcsb.org/pdb/explore.do?structureId=3LP1, DOI: DOI: 10.2210/pdb3lp1/pdb .Google ScholarThere is no corresponding record for this reference.
- 51Sastry, G. M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments J. Comput.-Aided Mol. Des. 2013, 27, 221– 234Google Scholar51Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichmentsSastry G Madhavi; Adzhigirey Matvey; Day Tyler; Annabhimoju Ramakrishna; Sherman WoodyJournal of computer-aided molecular design (2013), 27 (3), 221-34 ISSN:.Structure-based virtual screening plays an important role in drug discovery and complements other screening approaches. In general, protein crystal structures are prepared prior to docking in order to add hydrogen atoms, optimize hydrogen bonds, remove atomic clashes, and perform other operations that are not part of the x-ray crystal structure refinement process. In addition, ligands must be prepared to create 3-dimensional geometries, assign proper bond orders, and generate accessible tautomer and ionization states prior to virtual screening. While the prerequisite for proper system preparation is generally accepted in the field, an extensive study of the preparation steps and their effect on virtual screening enrichments has not been performed. In this work, we systematically explore each of the steps involved in preparing a system for virtual screening. We first explore a large number of parameters using the Glide validation set of 36 crystal structures and 1,000 decoys. We then apply a subset of protocols to the DUD database. We show that database enrichment is improved with proper preparation and that neglecting certain steps of the preparation process produces a systematic degradation in enrichments, which can be large for some targets. We provide examples illustrating the structural changes introduced by the preparation that impact database enrichment. While the work presented here was performed with the Protein Preparation Wizard and Glide, the insights and guidance are expected to be generalizable to structure-based virtual screening with other docking methods.
- 52(a) Schrödinger Release 2013-2: Schrödinger Suite 2013 Protein Preparation Wizard; Schrödinger, LLC: New York, 2013;Google ScholarThere is no corresponding record for this reference.(b) Schrödinger Release 2013-2: Epik, version 2.5; Schrödinger, LLC: New York, 2013;Google ScholarThere is no corresponding record for this reference.(c) Schrödinger Release 2013-2: Impact, version 6.0; Schrödinger, LLC: New York, 2013;Google ScholarThere is no corresponding record for this reference.(d) Schrödinger Release 2013-2: Prime, version 3; Schrödinger, LLC: New York, 2013.Google ScholarThere is no corresponding record for this reference.
- 53Jorgensen, W. L.; Maxwell, D. S.; TiradoRives, J. Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids J. Am. Chem. Soc. 1996, 118, 11225– 11236Google Scholar53Development and Testing of the OPLS All-Atom Force Field on Conformational Energetics and Properties of Organic LiquidsJorgensen, William L.; Maxwell, David S.; Tirado-Rives, JulianJournal of the American Chemical Society (1996), 118 (45), 11225-11236CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The parametrization and testing of the OPLS all-atom force field for org. mols. and peptides are described. Parameters for both torsional and nonbonded energetics have been derived, while the bond stretching and angle bending parameters have been adopted mostly from the AMBER all-atom force field. The torsional parameters were detd. by fitting to rotational energy profiles obtained from ab initio MO calcns. at the RHF/6-31G*//RHF/6-31G* level for more than 50 org. mols. and ions. The quality of the fits was high with av. errors for conformational energies of less than 0.2 kcal/mol. The force-field results for mol. structures are also demonstrated to closely match the ab initio predictions. The nonbonded parameters were developed in conjunction with Monte Carlo statistical mechanics simulations by computing thermodn. and structural properties for 34 pure org. liqs. including alkanes, alkenes, alcs., ethers, acetals, thiols, sulfides, disulfides, aldehydes, ketones, and amides. Av. errors in comparison with exptl. data are 2% for heats of vaporization and densities. The Monte Carlo simulations included sampling all internal and intermol. degrees of freedom. It is found that such non-polar and monofunctional systems do not show significant condensed-phase effects on internal energies in going from the gas phase to the pure liqs.
- 54Schrödinger Release 2013-2: Maestro, version 9.5; Schrödinger, LLC, New York, 2013.Google ScholarThere is no corresponding record for this reference.
- 55Schrödinger Release 2013-2: LigPrep, version 2.7; Schrödinger, LLC, New York, 2013.Google ScholarThere is no corresponding record for this reference.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 123 publications.
- Joseph P. Milton, Dorota Gryko. Photochemical Functionalization of 4-Diazoisoquinoline-1,3(2H,4H)-diones and Their 1-Sulfoxide Analogues. ACS Organic & Inorganic Au 2025, 5
(3)
, 205-210. https://doi.org/10.1021/acsorginorgau.5c00017
- Tao Zhang, Xinxin Ren, Jingkun Li, Yanhong Wang, Bin Wang, Ziren Chen, Yu Xia, Shaofeng Wu, Chenjiang Liu, Yonghong Zhang. Lewis Acidic Ionic-Liquid-Catalyzed Radical-Cascade Alkylation/Cyclization of N-Alkyl-N-methacryloyl Benzamides with Alkanes via Visible-Light-Induced Ligand-to-Metal Charge Transfer: Access to Alkylated Isoquinoline-1,3(2H,4H)-diones. Organic Letters 2024, 26
(48)
, 10259-10266. https://doi.org/10.1021/acs.orglett.4c03697
- Xinqiang Tan, Yuezhen Li, Ziyou Hao, Jia Wang, Xiangqian Liu, Bingqing Liu, Jianmei Yuan, Lizhen Fang, Ping-Xin Zhou, Yingling Wang. Pentafluorosulfanylation of Acrylamides: The Synthesis of SF5-Containing Isoquinolinediones with SF5Cl. The Journal of Organic Chemistry 2024, 89
(21)
, 15941-15952. https://doi.org/10.1021/acs.joc.4c02181
- Xinxin Ren, Tao Zhang, Bin Wang, Weiwei Jin, Yu Xia, Shaofeng Wu, Chenjiang Liu, Yonghong Zhang. Visible-Light-Driven Bifunctional Photocatalytic Radical-Cascade Selenocyanation/Cyclization of Acrylamides with KSeCN. The Journal of Organic Chemistry 2024, 89
(8)
, 5783-5796. https://doi.org/10.1021/acs.joc.4c00362
- Wenjuan Zhang, Yaqi Song, Tian-Yu Sun, Dawei Wang, Xiao-Feng Xia. Photocatalytic Proton-Coupled Electron Transfer Enabled Radical Cyclization for Isoquinoline-1,3-diones Synthesis. The Journal of Organic Chemistry 2024, 89
(7)
, 5060-5068. https://doi.org/10.1021/acs.joc.4c00282
- Vishal Jyoti Roy, Neha Dagar, Swagata Choudhury, Sudipta Raha Roy. Unified Approach to Diverse Heterocyclic Synthesis: Organo-Photocatalyzed Carboacylation of Alkenes and Alkynes from Feedstock Aldehydes and Alcohols. The Journal of Organic Chemistry 2023, 88
(21)
, 15374-15388. https://doi.org/10.1021/acs.joc.3c01884
- Zeguo Fang, Wen Liu, Nawaf Al-Maharik, Ruizhe Cao, Yingxue Huang, Yiting Yuan, Qian Zhang, Dong Li. Silver-Catalyzed Cascade Radical Bicyclization Reaction: An Atom- and Step-Economical Strategy Accessing γ-Lactam Containing Isoquinolinediones. The Journal of Organic Chemistry 2023, 88
(21)
, 15428-15436. https://doi.org/10.1021/acs.joc.3c01964
- Ajit Dhananjay Jagtap, Robert J. Geraghty, Zhengqiang Wang. Inhibiting HCMV pUL89-C Endonuclease with Metal-Binding Compounds. Journal of Medicinal Chemistry 2023, 66
(20)
, 13874-13887. https://doi.org/10.1021/acs.jmedchem.3c01280
- Tianyu He, Tiffany C. Edwards, Jiashu Xie, Hideki Aihara, Robert J. Geraghty, Zhengqiang Wang. 4,5-Dihydroxypyrimidine Methyl Carboxylates, Carboxylic Acids, and Carboxamides as Inhibitors of Human Cytomegalovirus pUL89 Endonuclease. Journal of Medicinal Chemistry 2022, 65
(7)
, 5830-5849. https://doi.org/10.1021/acs.jmedchem.2c00203
- Lanjun Su, Pan Xue, Xin Zhu, Huan Sun, Jikai Liu, Chengming Wang. Transition-Metal- and Light-Free Generation of an Iminyl Radical: Facile Approach to Oxindoles and Isoquinolinediones with a Quaternary Carbon Center via Cyanoalkylarylation. The Journal of Organic Chemistry 2022, 87
(1)
, 874-883. https://doi.org/10.1021/acs.joc.1c02593
- Xiaoshen Ma, Charles S. Yeung. Achieving C(sp2)–C(sp3) Coupling with BCP-F2 Building Blocks via Barluenga Coupling: A Comparative Approach. The Journal of Organic Chemistry 2021, 86
(15)
, 10672-10698. https://doi.org/10.1021/acs.joc.1c01370
- Antonella Messore, Angela Corona, Valentina Noemi Madia, Francesco Saccoliti, Valeria Tudino, Alessandro De Leo, Davide Ialongo, Luigi Scipione, Daniela De Vita, Giorgio Amendola, Ettore Novellino, Sandro Cosconati, Mathieu Métifiot, Marie-Line Andreola, Francesca Esposito, Nicole Grandi, Enzo Tramontano, Roberta Costi, Roberto Di Santo. Quinolinonyl Non-Diketo Acid Derivatives as Inhibitors of HIV-1 Ribonuclease H and Polymerase Functions of Reverse Transcriptase. Journal of Medicinal Chemistry 2021, 64
(12)
, 8579-8598. https://doi.org/10.1021/acs.jmedchem.1c00535
- Maria E. Cilento, Karen A. Kirby, Stefan G. Sarafianos. Avoiding Drug Resistance in HIV Reverse Transcriptase. Chemical Reviews 2021, 121
(6)
, 3271-3296. https://doi.org/10.1021/acs.chemrev.0c00967
- Zhenxing Zhang, Chunqi Jia, Xiangtao Kong, Muzammal Hussain, Zhenyu Liu, Wanqing Liang, Ling Jiang, Hao Jiang, Junyan Ma. Sustainable Approach to Azaheterocyclic Acetamides by Decarboxylative Aminoformylation. ACS Sustainable Chemistry & Engineering 2020, 8
(44)
, 16463-16468. https://doi.org/10.1021/acssuschemeng.0c05118
- Lei Wang, Stefan G. Sarafianos, Zhengqiang Wang. Cutting into the Substrate Dominance: Pharmacophore and Structure-Based Approaches toward Inhibiting Human Immunodeficiency Virus Reverse Transcriptase-Associated Ribonuclease H. Accounts of Chemical Research 2020, 53
(1)
, 218-230. https://doi.org/10.1021/acs.accounts.9b00450
- Chao Dong, Xin Wang, Zibo Pei, Ruwei Shen. Metal-Free Denitrogenative C–C Couplings of Pyridotriazoles with Boronic Acids To Afford α-Secondary and α-Tertiary Pyridines. Organic Letters 2019, 21
(11)
, 4148-4152. https://doi.org/10.1021/acs.orglett.9b01334
- Yuanyong Yang, Yingxian Li, Cheng Cheng, Guo Yang, Shuiying Wan, Jiquan Zhang, Yuanhu Mao, Yonglong Zhao, Lin Zhang, Chun Li, Lei Tang. Reductant-Free Aerobic Hydroxylation of Isoquinoline-1,3(2H,4H)-dione Derivatives. The Journal of Organic Chemistry 2019, 84
(4)
, 2316-2324. https://doi.org/10.1021/acs.joc.8b02977
- Masato Ohsumi and Nagatoshi Nishiwaki . Selective Synthesis of (Benzyl)biphenyls by Successive Suzuki–Miyaura Coupling of Phenylboronic Acids with 4-Bromobenzyl Acetate under Air Atmosphere. ACS Omega 2017, 2
(11)
, 7767-7771. https://doi.org/10.1021/acsomega.7b01450
- Aaron Wegener and Kenneth A. Miller . Total Synthesis of Avrainvilleol. The Journal of Organic Chemistry 2017, 82
(21)
, 11655-11658. https://doi.org/10.1021/acs.joc.7b02028
- Sanjeev Kumar V. Vernekar, Jing Tang, Bulan Wu, Andrew D. Huber, Mary C. Casey, Nataliya Myshakina, Daniel J. Wilson, Jayakanth Kankanala, Karen A. Kirby, Michael A. Parniak, Stefan G. Sarafianos, and Zhengqiang Wang . Double-Winged 3-Hydroxypyrimidine-2,4-diones: Potent and Selective Inhibition against HIV-1 RNase H with Significant Antiviral Activity. Journal of Medicinal Chemistry 2017, 60
(12)
, 5045-5056. https://doi.org/10.1021/acs.jmedchem.7b00440
- Han Ju, Jian Zhang, Boshi Huang, Dongwei Kang, Bing Huang, Xinyong Liu, and Peng Zhan . Inhibitors of Influenza Virus Polymerase Acidic (PA) Endonuclease: Contemporary Developments and Perspectives. Journal of Medicinal Chemistry 2017, 60
(9)
, 3533-3551. https://doi.org/10.1021/acs.jmedchem.6b01227
- Xiao-Feng Xia, Su-Li Zhu, Jian-Biao Liu, Dawei Wang, and Yong-Min Liang . Copper-Catalyzed Radical-Promoted Aminocyclization of Acrylamides with N-Fluorobenzenesulfonimide. The Journal of Organic Chemistry 2016, 81
(24)
, 12482-12488. https://doi.org/10.1021/acs.joc.6b02337
- Bulan Wu, Jing Tang, Daniel J. Wilson, Andrew D. Huber, Mary C. Casey, Juan Ji, Jayakanth Kankanala, Jiashu Xie, Stefan G. Sarafianos, and Zhengqiang Wang . 3-Hydroxypyrimidine-2,4-dione-5-N-benzylcarboxamides Potently Inhibit HIV-1 Integrase and RNase H. Journal of Medicinal Chemistry 2016, 59
(13)
, 6136-6148. https://doi.org/10.1021/acs.jmedchem.6b00040
- Jayakanth Kankanala, Karen A. Kirby, Feng Liu, Lena Miller, Eva Nagy, Daniel J. Wilson, Michael A. Parniak, Stefan G. Sarafianos, and Zhengqiang Wang . Design, Synthesis, and Biological Evaluations of Hydroxypyridonecarboxylic Acids as Inhibitors of HIV Reverse Transcriptase Associated RNase H. Journal of Medicinal Chemistry 2016, 59
(10)
, 5051-5062. https://doi.org/10.1021/acs.jmedchem.6b00465
- Peng Zhan, Christophe Pannecouque, Erik De Clercq, and Xinyong Liu . Anti-HIV Drug Discovery and Development: Current Innovations and Future Trends. Journal of Medicinal Chemistry 2016, 59
(7)
, 2849-2878. https://doi.org/10.1021/acs.jmedchem.5b00497
- Jing Tang, Feng Liu, Eva Nagy, Lena Miller, Karen A. Kirby, Daniel J. Wilson, Bulan Wu, Stefan G. Sarafianos, Michael A. Parniak, and Zhengqiang Wang . 3-Hydroxypyrimidine-2,4-diones as Selective Active Site Inhibitors of HIV Reverse Transcriptase-Associated RNase H: Design, Synthesis, and Biochemical Evaluations. Journal of Medicinal Chemistry 2016, 59
(6)
, 2648-2659. https://doi.org/10.1021/acs.jmedchem.5b01879
- Shi Tang, You-Lin Deng, Jie Li, Wen-Xin Wang, Guo-Liang Ding, Ming-Wei Wang, Zhu-Ping Xiao, Ying-Chun Wang, and Rui-Long Sheng . Synthesis of Perfluorinated Isoquinolinediones through Visible-Light-Induced Cyclization of Alkenes. The Journal of Organic Chemistry 2015, 80
(24)
, 12599-12605. https://doi.org/10.1021/acs.joc.5b01803
- Jubi John, Youngju Kim, Nicholas Bennett, Kalyan Das, Sandra Liekens, Lieve Naesens, Eddy Arnold, Anita R. Maguire, Matthias Götte, Wim Dehaen, and Jan Balzarini . Pronounced Inhibition Shift from HIV Reverse Transcriptase to Herpetic DNA Polymerases by Increasing the Flexibility of α-Carboxy Nucleoside Phosphonates. Journal of Medicinal Chemistry 2015, 58
(20)
, 8110-8127. https://doi.org/10.1021/acs.jmedchem.5b01180
- Wei Niu, Yanlong Liu, Erling Dai, Jiarui Sun, Yuyu Dai, Xiangsheng Xu. Synthesis of amide-functionalized isoquinoline derivatives by photo-induced carbamoyl radical cascade amidation/cyclization. Organic & Biomolecular Chemistry 2025, 23
(23)
, 5533-5541. https://doi.org/10.1039/D5OB00465A
- Saikat Pandit, Tobias Ritter. Copper‐Photoredox‐Catalyzed C(sp
3
)–C(sp
3
) Reductive Cross‐Coupling of Alkyl Bromides with BCP‐Thianthrenium Reagents. Angewandte Chemie 2025, 55 https://doi.org/10.1002/ange.202506785
- Saikat Pandit, Tobias Ritter. Copper‐Photoredox‐Catalyzed C(sp
3
)–C(sp
3
) Reductive Cross‐Coupling of Alkyl Bromides with BCP‐Thianthrenium Reagents. Angewandte Chemie International Edition 2025, 55 https://doi.org/10.1002/anie.202506785
- Ren‐Rui Xu, Chang‐Sheng Kuai, Xiao‐Feng Wu. Palladium‐Catalyzed Selectivity Controlled Highly Efficient Carbonylative Synthesis of Isoquinoline‐1,3(2
H
,4
H
)‐diones and Indanones. ChemistryEurope 2025, 3
(2)
https://doi.org/10.1002/ceur.202400105
- Lei Huang, Chaodong Wang, Zhi Chen, Qianxi Jin, Shengjie Song, Jiadi Zhou, Jianjun Li. Photoinduced EDA Complex‐Initiated Synthesis of Fluoroalkylated Isoquinolinonediones. Chemistry – A European Journal 2025, 31
(3)
https://doi.org/10.1002/chem.202403286
- Fathi Shalbi, Ahmed R. Ali. A mini-review on integrase inhibitors: The cornerstone of next-generation HIV treatment. European Journal of Medicinal Chemistry 2024, 279 , 116900. https://doi.org/10.1016/j.ejmech.2024.116900
- An-Qi Miao, Yu-Xin Wang, Lin-Lin Xu, Wen-Juan Hao, Shu-Jiang Tu, Jin-Peng Zhang, Bo Jiang. Pd(II)/N,N′-Disulfonyl bisimidazoline-catalyzed asymmetric arylation of isoquinoline-1,3,4-trione-derived ketimines. Green Synthesis and Catalysis 2024, 5
(3)
, 165-169. https://doi.org/10.1016/j.gresc.2023.07.001
- Lei Huang, Jun Sun, Boxuan Sun, Shengjie Song, Jianjun Li. Regioselective synthesis of isoquinolinonediones through remote unactivated C(sp
3
)–H bonds. Chemical Communications 2024, 60
(36)
, 4818-4821. https://doi.org/10.1039/D4CC00916A
- Dong-Liang Zhang, Zhang-Gao Le, Qing Li, Zong-Bo Xie, Wen-Wen Yang, Zhi-Qiang Zhu. Visible-light-driven EDA complex-promoted cascade cyclization to construct 4-cyanoalkyl isoquinoline-1,3-diones. Chemical Communications 2024, 60
(21)
, 2958-2961. https://doi.org/10.1039/D4CC00092G
- Hongsheng Ma, Tongyan Yu, Siliang You, Ming‐Zhi Zhang, Chao Deng. Synthesis of nitro‐containing isoquinoline‐1,3‐diones with tert‐butyl nitrite. Journal of Heterocyclic Chemistry 2024, 61
(2)
, 331-335. https://doi.org/10.1002/jhet.4762
- Qing Li, Zhi-Qiang Zhu, Wen-Yi Zhang, Zhang-Gao Le, Zong-Bo Xie. Visible-light-induced decarboxylative cascade cyclization of acryloylbenzamides with
N
-hydroxyphthalimide esters
via
EDA complexes. Organic & Biomolecular Chemistry 2024, 22
(5)
, 965-969. https://doi.org/10.1039/D3OB01970E
- Jinli Xu, Chenyue Zhao, Dongping Cheng, Xiaoliang Xu. Synthesis of Isoquinolinediones via the Radical Addition/Cyclization of N‐Alkyl‐N‐methacryloylbenzamides with Monoalkyl Oxalates. Advanced Synthesis & Catalysis 2023, 365
(24)
, 4527-4532. https://doi.org/10.1002/adsc.202301223
- Archana Vijayakumar, M. Manod, R. Bharath Krishna, Abra Mathew, Chithra Mohan. Diversely functionalized isoquinolines and their core-embedded heterocyclic frameworks: a privileged scaffold for medicinal chemistry. RSC Medicinal Chemistry 2023, 14
(12)
, 2509-2534. https://doi.org/10.1039/D3MD00248A
- Dharmraj V. Pathak, Sneha R. Sagar, Hardik G. Bhatt, Paresh K. Patel. A search for potential anti-HIV phytoconstituents from the natural product repository. Advances in Traditional Medicine 2023, 23
(4)
, 953-984. https://doi.org/10.1007/s13596-022-00646-2
- Pengfei Jia, Zhiqian Lin, Cankun Luo, Jiao Liang, Ruizhi Lai, Li Guo, Yuan Yao, Yong Wu. Synthesis of spirotriazolines and spirooxadiazolines
via
light-induced 1,3-dipolar [3+2] cycloadditions. New Journal of Chemistry 2023, 47
(44)
, 20248-20252. https://doi.org/10.1039/D3NJ03037G
- Xiao-Yu Lu, Fu-Yi Shui, Xiang Zhang, Rui Huang, Zi-Zhen Wang, Yi-Wei Song, Fan Jiang, Gui-Xian Yang, Yan-Xi Sun. Palladium catalyzed tandem cyclization of acryloylbenzamides and
N
-arylacrylamides with epoxides: access to functionalized isoquinolinediones and oxindoles. Organic Chemistry Frontiers 2023, 10
(22)
, 5616-5622. https://doi.org/10.1039/D3QO01243C
- Shujing Xu, Lin Sun, Xinyong Liu, Peng Zhan. Opportunities and challenges in new HIV therapeutic discovery: what is the next step?. Expert Opinion on Drug Discovery 2023, 18
(11)
, 1195-1199. https://doi.org/10.1080/17460441.2023.2246872
- Fatma Mohamed A. Abid, Nurul Syahirah Salim, Roswanira Abdul Wahab, Siti Ernieyanti Hashim, Murugesan Sankaranarayanan, Ajmal Bhat, Joazaizulfazli Jamalis. An Overview of the Anti-HIV Potential of Thiazolidinone Derivatives. Anti-Infective Agents 2023, 21
(5)
https://doi.org/10.2174/2211352521666230718094413
- Cai Zhang, Demao Chen, Jie-Ping Wan, Yunyun Liu. Recent Advances in Electrochemical Cascade Cyclization Reactions. Synthesis 2023, 55
(18)
, 2911-2925. https://doi.org/10.1055/a-2039-1728
- Yushen Yu, Zhihui Liang, Lele Zhang, Yuhan Lin, Min Liu, Shanshan Wang, Mingqiang Huang, Lina Cai, Shunyou Cai. Photocatalytic dehydrogenative oxidation/amination of 2-alkyl benzamides under transition-metal-free conditions. Organic Chemistry Frontiers 2023, 10
(17)
, 4400-4407. https://doi.org/10.1039/D3QO00878A
- Dimitrios Moianos, Georgia-Myrto Prifti, Maria Makri, Grigoris Zoidis. Targeting Metalloenzymes: The “Achilles’ Heel” of Viruses and Parasites. Pharmaceuticals 2023, 16
(6)
, 901. https://doi.org/10.3390/ph16060901
- Jia-Xiong Kang, Guang-Kuan Zhao, Xiu-Ming Yang, Mou-Xin Huang, Wen-Qi Hui, Rong Zeng, Qin Ouyang. Recent advances on dual inhibitors targeting HIV reverse transcriptase associated polymerase and ribonuclease H. European Journal of Medicinal Chemistry 2023, 250 , 115196. https://doi.org/10.1016/j.ejmech.2023.115196
- Zhuang-Zhi Shi, Tongyan Yu, Hongsheng Ma, Long-Xiao Chi, Siliang You, Chao Deng. Recent advances in radical cascade cyclization of 1,n-enynes with trifluoromethylating agents. Tetrahedron 2023, 131 , 133216. https://doi.org/10.1016/j.tet.2022.133216
- Youchun Huang, Ke Kang, Xiuwen Zhong, Yongping Yu, Wenteng Chen. Rh(III)-catalyzed C H activation/intramolecular annulation for the synthesis of N-methoxydihydropyrimidin-2-one fused heterocycles. Tetrahedron Letters 2023, 115 , 154311. https://doi.org/10.1016/j.tetlet.2022.154311
- Tianyu He, Tiffany C. Edwards, Ryuichi Majima, Eunkyung Jung, Jayakanth Kankanala, Jiashu Xie, Robert J. Geraghty, Zhengqiang Wang. Repurposing N-hydroxy thienopyrimidine-2,4-diones (HtPD) as inhibitors of human cytomegalovirus pUL89 endonuclease: Synthesis and biological characterization. Bioorganic Chemistry 2022, 129 , 106198. https://doi.org/10.1016/j.bioorg.2022.106198
- Lina Zhang, Fenju Wei, Jiwei Zhang, Chuanfeng Liu, Nerea López-Carrobles, Xinyong Liu, Luis Menéndez-Arias, Peng Zhan. Current medicinal chemistry strategies in the discovery of novel HIV-1 ribonuclease H inhibitors. European Journal of Medicinal Chemistry 2022, 243 , 114760. https://doi.org/10.1016/j.ejmech.2022.114760
- D. Wen, Z. Shen, X. Qi, X.‐F. Wu. Palladium‐Catalyzed Cascade Carbonylative Synthesis of Functionalized Isoquinoline‐1,3‐diones and Oxindoles Using Dimethyl Carbonate as both Solvent and Reactant. European Journal of Organic Chemistry 2022, 2022
(42)
https://doi.org/10.1002/ejoc.202200971
- Jianyi Shi, Zheng Zeng, Shengting Xu, Zechun Cai, Yuehua Luo, Yongbo Fan, Zhongzhi Zhu, Tingting Wen, Xiuwen Chen. Cross-coupling of 2-methylquinolines and in-situ activated isoquinolines: Construction of 1,2-disubstituted isoquinolinones. Chinese Chemical Letters 2022, 33
(11)
, 4874-4877. https://doi.org/10.1016/j.cclet.2022.02.032
- Yan-Ning Niu, Xiao-Feng Xia. Recent developments in the synthesis of the isoquinoline-1,3(2
H
,4
H
)-dione by radical cascade reaction. Organic & Biomolecular Chemistry 2022, 20
(40)
, 7861-7885. https://doi.org/10.1039/D2OB01554D
- Zhihui Liang, Chao Liu, Jiajun Fan, Mingze Wang, Xuanyu Yan, Mingqiang Huang, Shunyou Cai. Photocatalytic dehydrogenated etherification of 2-aryl benzylic alcohols. Green Chemistry 2022, 24
(19)
, 7442-7447. https://doi.org/10.1039/D2GC02030K
- Lina Zhang, Fenju Wei, David Borrego, Fabao Zhao, Javier Martínez del Río, Estrella Frutos-Beltrán, Jiwei Zhang, Shujing Xu, Nerea López-Carrobles, Shenghua Gao, Dongwei Kang, Christophe Pannecouque, Erik De Clercq, Xinyong Liu, Luis Menéndez-Arias, Peng Zhan. Design, synthesis, and biological evaluation of novel double-winged galloyl derivatives as HIV-1 RNase H inhibitors. European Journal of Medicinal Chemistry 2022, 240 , 114563. https://doi.org/10.1016/j.ejmech.2022.114563
- Eunkyung Jung, Ryuichi Majima, Tiffany C. Edwards, Ruben Soto‐Acosta, Robert J. Geraghty, Zhengqiang Wang. 8‐Hydroxy‐1,6‐naphthyridine‐7‐carboxamides as Inhibitors of Human Cytomegalovirus pUL89 Endonuclease. ChemMedChem 2022, 17
(17)
https://doi.org/10.1002/cmdc.202200334
- Diancong Chen, Haiping Yang, Zhong Li, Peter Maienfisch, Xiaoyong Xu. Design, synthesis and nematicidal activities of trifluorobutene hydroxamic acid derivatives against Meloidogyne incognita. Journal of Molecular Structure 2022, 1264 , 133191. https://doi.org/10.1016/j.molstruc.2022.133191
- Xingxing Cai, Jiahui Fu, Li Gu, Dongping Cheng, Hong Wang, Xiaoliang Xu. Visible‐light‐promoted Cascade Reaction of Acryloylbenzamides with Carboxylic Acids: Metal‐free Synthesis of Isoquinolinediones. Asian Journal of Organic Chemistry 2022, 11
(8)
https://doi.org/10.1002/ajoc.202200318
- Shidi Ma, Pengsheng Zhou, Xu Fan, Dianjun Li, Jinhui Yang. Base-promoted cascade radical difluoroalkylation/cyclization of acrylamides for the synthesis of CF2-containing oxindoles and isoquinoline-1,3-diones. Tetrahedron Letters 2022, 102 , 153933. https://doi.org/10.1016/j.tetlet.2022.153933
- Jiahui Fu, Xingxing Cai, Yihuo Liu, Jinghua Li, Dongping Cheng, Xiaoliang Xu. Synthesis of Isoquinoline-1,3(2H,4H)-diones by Visible-Light-Mediated Cyclization of Acryloylbenzamides with Alkylboronic Acids, Arylsulfonyl Hydrazides and Oxime Esters. Synthesis 2022, 54
(11)
, 2707-2723. https://doi.org/10.1055/a-1734-9782
- Xingxing Cai, Yihuo Liu, Siyu Ding, Jiahui Fu, Jinghua Li, Dongping Cheng, Xiaoliang Xu. Visible light-induced radical cascade reaction of acryloylbenzamides with N-hydroxyphthalimide esters. Tetrahedron Letters 2022, 95 , 153722. https://doi.org/10.1016/j.tetlet.2022.153722
- Yameng Wan, Qingfeng Liu, Hao Wu, Zhiguo Zhang, Guisheng Zhang. 2,11-Dimethoxyldipyridopurinone as an efficient reducing visible-light photocatalyst for organic transformations. Organic Chemistry Frontiers 2022, 9
(6)
, 1634-1641. https://doi.org/10.1039/D1QO01914G
- Lei Wang, Tiffany C. Edwards, Rajkumar Lalji Sahani, Jiashu Xie, Hideki Aihara, Robert J. Geraghty, Zhengqiang Wang. Metal binding 6-arylthio-3-hydroxypyrimidine-2,4-diones inhibited human cytomegalovirus by targeting the pUL89 endonuclease of the terminase complex. European Journal of Medicinal Chemistry 2021, 222 , 113640. https://doi.org/10.1016/j.ejmech.2021.113640
- Jianlian Liu, Chaojun Du, Hong Hao. Crystal structure of 4-(2,2-difluoroethyl)-2,4-dimethyl-6-(trifluoromethyl)isoquinoline-1,3(2
H
,4
H
)-dione, C
14
H
12
F
5
NO
2. Zeitschrift für Kristallographie - New Crystal Structures 2021, 236
(5)
, 979-980. https://doi.org/10.1515/ncrs-2021-0179
- Sandeep Kumar, Tariq A. Shah, Tharmalingam Punniyamurthy. Recent advances in the application of tetrabromomethane in organic synthesis. Organic Chemistry Frontiers 2021, 8
(15)
, 4288-4314. https://doi.org/10.1039/D0QO01369B
- Ying‐Xian Li, Xiao‐Bing Feng, Xue‐Min Jia, Huang Jin, Fei Chen, Yong‐Long Zhao, Ji‐Quan Zhang, Jian‐Ta Wang, Bing Guo, Lei Tang, Yuan‐Yong Yang. Regiodivergent Functionalization of Isoquinoline‐1,3(2
H
,4
H
)‐dione Derivatives via Aerobic Umpolung. Asian Journal of Organic Chemistry 2021, 10
(6)
, 1508-1513. https://doi.org/10.1002/ajoc.202100194
- Huixing Li, Xinlong Han, Bin Cao. Crystal structure of 4-(2,2-difluoroethyl)-2,4,6-trimethylisoquinoline-1,3(2
H
,4
H
)-dione, C
14
H
15
F
2
NO
2. Zeitschrift für Kristallographie - New Crystal Structures 2021, 236
(3)
, 499-501. https://doi.org/10.1515/ncrs-2020-0635
- Yue Ma, Estrella Frutos-Beltrán, Dongwei Kang, Christophe Pannecouque, Erik De Clercq, Luis Menéndez-Arias, Xinyong Liu, Peng Zhan. Medicinal chemistry strategies for discovering antivirals effective against drug-resistant viruses. Chemical Society Reviews 2021, 50
(7)
, 4514-4540. https://doi.org/10.1039/D0CS01084G
- Ren‐You Guan, Chan‐Wei Hsu, Jeng‐Liang Han. Trifluoroethanol‐mediated Decarboxylative Addition Reactions of β‐Ketoacids with Diverse Electrophiles. Asian Journal of Organic Chemistry 2021, 10
(1)
, 273-284. https://doi.org/10.1002/ajoc.202000553
- Fenju Wei, Edeildo Ferreira da Silva-Júnior, Xinyong Liu, Peng Zhan. HIV-1 and HBV RNase H as Metal-Chelating Inhibitors: Discovery and Medicinal Chemistry Strategies. 2021, 585-602. https://doi.org/10.1007/978-3-030-71165-8_28
- Jiaxuan Shao, Youjia Wang, Silu Chen, Shaowei Wang, Yanhui Wu, Xulian Li, Weihua Zhang, Chao Deng. Synthesis of CF2H-containing isoquinoline-1,3-diones through metal-free, visible-light and air-promoted radical difluoromethylation/cyclization of N-benzamides. Tetrahedron 2021, 79 , 131822. https://doi.org/10.1016/j.tet.2020.131822
- Cheng Cheng, Ying-Xian Li, Xue-Min Jia, Ji-Quan Zhang, Yong-Long Zhao, Wei Feng, Lei Tang, Yuan-Yong Yang. Enantioselective amination of 4-alkylisoquinoline-1,3(2
H
,4
H
)-dione derivatives. RSC Advances 2020, 10
(70)
, 42912-42915. https://doi.org/10.1039/D0RA07806A
- Fenju Wei, Dongwei Kang, Luis Menéndez‐Arias, Xinyong Liu, Peng Zhan. Recent Developments in Small Molecular HIV‐1 and Hepatitis B Virus RNase H Inhibitors. 2020, 273-292. https://doi.org/10.1002/9781119640868.ch9
- Yingpeng Su, Rong Zhang, Wenxuan Xue, Xuan Liu, Yanan Zhao, Ke-Hu Wang, Danfeng Huang, Congde Huo, Yulai Hu. Visible-light-promoted acyl radical cascade reaction for accessing acylated isoquinoline-1,3(2
H
,4
H
)-dione derivatives. Organic & Biomolecular Chemistry 2020, 18
(10)
, 1940-1948. https://doi.org/10.1039/D0OB00086H
- Yali Sang, Christophe Pannecouque, Erik De Clercq, Chunlin Zhuang, Fener Chen. Pharmacophore-fusing design of pyrimidine sulfonylacetanilides as potent non-nucleoside inhibitors of HIV-1 reverse transcriptase. Bioorganic Chemistry 2020, 96 , 103595. https://doi.org/10.1016/j.bioorg.2020.103595
- Enzo Tramontano, Angela Corona, Luis Menéndez-Arias. Ribonuclease H, an unexploited target for antiviral intervention against HIV and hepatitis B virus. Antiviral Research 2019, 171 , 104613. https://doi.org/10.1016/j.antiviral.2019.104613
- Yanfei Liu, Jiaping Wu, Baiyang Qian, Yongjia Shang. Controllable construction of isoquinolinedione and isocoumarin scaffolds
via
Rh
III
-catalyzed C–H annulation of
N
-tosylbenzamides with diazo compounds. Organic & Biomolecular Chemistry 2019, 17
(38)
, 8768-8777. https://doi.org/10.1039/C9OB01789E
- Shaowei Wang, Peng Dai, Zhichao Yan, Youjia Wang, Jiaxuan Shao, Yanhui Wu, Chao Deng, Weihua Zhang. Metal‐free, Visible‐Light‐Induced Radical Trifluoromethylation/Cyclization of N‐benzamides with CF
3
SO
2
Na to Synthesize CF
3
‐Containing Isoquinoline‐1,3‐diones. ChemistrySelect 2019, 4
(35)
, 10329-10333. https://doi.org/10.1002/slct.201902545
- Maojian Lu, Tao Zhang, Dabao Tan, Chengzhu Chen, Ying Zhang, Mingqiang Huang, Shunyou Cai. Visible‐Light‐Promoted Oxidative Alkylarylation of
N
‐Aryl/Benzoyl Acrylamides Through Direct C−H Bond Functionalization. Advanced Synthesis & Catalysis 2019, 361
(18)
, 4237-4242. https://doi.org/10.1002/adsc.201900712
- Yali Sang, Sheng Han, Shuwen Han, Christophe Pannecouque, Erik De Clercq, Chunlin Zhuang, Fener Chen. Follow on-based optimization of the biphenyl-DAPYs as HIV-1 nonnucleoside reverse transcriptase inhibitors against the wild-type and mutant strains. Bioorganic Chemistry 2019, 89 , 102974. https://doi.org/10.1016/j.bioorg.2019.102974
- Ross D. Overacker, Somdev Banerjee, George F. Neuhaus, Selena Milicevic Sephton, Alexander Herrmann, James A. Strother, Ruth Brack-Werner, Paul R. Blakemore, Sandra Loesgen. Biological evaluation of molecules of the azaBINOL class as antiviral agents: Inhibition of HIV-1 RNase H activity by 7-isopropoxy-8-(naphth-1-yl)quinoline. Bioorganic & Medicinal Chemistry 2019, 27
(16)
, 3595-3604. https://doi.org/10.1016/j.bmc.2019.06.044
- Yong You, Wen-Ya Lu, Ke-Xin Xie, Jian-Qiang Zhao, Zhen-Hua Wang, Wei-Cheng Yuan. Enantioselective synthesis of isoquinoline-1,3(2
H
,4
H
)-dione derivatives
via
a chiral phosphoric acid catalyzed aza-Friedel–Crafts reaction. Chemical Communications 2019, 55
(58)
, 8478-8481. https://doi.org/10.1039/C9CC04057A
- Antonia Di Mola, Consiglia Tedesco, Antonio Massa. Metal-Free Air Oxidation in a Convenient Cascade Approach for the Access to Isoquinoline-1,3,4(2H)-triones. Molecules 2019, 24
(11)
, 2177. https://doi.org/10.3390/molecules24112177
- Yu-Jue Chen, Yan-Hong He, Zhi Guan. Metal-free visible-light-promoted thiocyanation/cyclization cascade for the synthesis of thiocyanato-containing isoquinolinediones. Tetrahedron 2019, 75
(22)
, 3053-3061. https://doi.org/10.1016/j.tet.2019.04.053
- Ping Gao, Xueshun Wang, Lin Sun, Xiqiang Cheng, Vasanthanathan Poongavanam, Jacob Kongsted, Mar Álvarez, Joanna Luczkowiak, Christophe Pannecouque, Erik De Clercq, Kuo‐Hsiung Lee, Chin‐Ho Chen, Huiqing Liu, Luis Menéndez‐Arias, Xinyong Liu, Peng Zhan. Design, synthesis, and biologic evaluation of novel galloyl derivatives as
HIV
‐1
RN
ase H inhibitors. Chemical Biology & Drug Design 2019, 93
(4)
, 582-589. https://doi.org/10.1111/cbdd.13455
- Jing Tang, Ha T. Do, Andrew D. Huber, Mary C. Casey, Karen A. Kirby, Daniel J. Wilson, Jayakanth Kankanala, Michael A. Parniak, Stefan G. Sarafianos, Zhengqiang Wang. Pharmacophore-based design of novel 3-hydroxypyrimidine-2,4-dione subtypes as inhibitors of HIV reverse transcriptase-associated RNase H: Tolerance of a nonflexible linker. European Journal of Medicinal Chemistry 2019, 166 , 390-399. https://doi.org/10.1016/j.ejmech.2019.01.081
- Tiexin Zhang, Xiangyang Guo, Yusheng Shi, Cheng He, Chunying Duan. Dye-incorporated coordination polymers for direct photocatalytic trifluoromethylation of aromatics at metabolically susceptible positions. Nature Communications 2018, 9
(1)
https://doi.org/10.1038/s41467-018-05919-6
- Tiexin Zhang, Yusheng Shi, Sen Zhang, Chen Jia, Cheng He, Chunying Duan. Thiophene insertion for continuous modulation of the photoelectronic properties of triphenylamine-based metal–organic frameworks for photocatalytic sulfonylation–cyclisation of activated alkenes. New Journal of Chemistry 2018, 42
(22)
, 18448-18457. https://doi.org/10.1039/C8NJ04151B
- Zeng-Zeng Li, Jian Yu, Liang-Neng Wang, Shi-Lu Chen, Rui-Long Sheng, Shi Tang. Cascade radical cyclization/cross-coupling of halobenzamides by synergistic Cu/Fe catalysis: An access to 7-tert-alkylated isoquinolinediones. Tetrahedron 2018, 74
(45)
, 6558-6568. https://doi.org/10.1016/j.tet.2018.09.036
- Songhai Huang, Pengfei Niu, Yingpeng Su, Dongcheng Hu, Congde Huo. Tandem radical cyclization of
N
-methacryloyl benzamides with CBr
4
to construct brominated isoquinolinediones. Organic & Biomolecular Chemistry 2018, 16
(41)
, 7748-7752. https://doi.org/10.1039/C8OB01964A
- Lei Wang, Jing Tang, Andrew D. Huber, Mary C. Casey, Karen A. Kirby, Daniel J. Wilson, Jayakanth Kankanala, Michael A. Parniak, Stefan G. Sarafianos, Zhengqiang Wang. 6-Biphenylmethyl-3-hydroxypyrimidine-2,4-diones potently and selectively inhibited HIV reverse transcriptase-associated RNase H. European Journal of Medicinal Chemistry 2018, 156 , 680-691. https://doi.org/10.1016/j.ejmech.2018.07.035
- Lei Wang, Jing Tang, Andrew D. Huber, Mary C. Casey, Karen A. Kirby, Daniel J. Wilson, Jayakanth Kankanala, Jiashu Xie, Michael A. Parniak, Stefan G. Sarafianos, Zhengqiang Wang. 6-Arylthio-3-hydroxypyrimidine-2,4-diones potently inhibited HIV reverse transcriptase-associated RNase H with antiviral activity. European Journal of Medicinal Chemistry 2018, 156 , 652-665. https://doi.org/10.1016/j.ejmech.2018.07.039
- Lin Sun, Ping Gao, Guanyu Dong, Xujie Zhang, Xiqiang Cheng, Xiao Ding, Xueshun Wang, Dirk Daelemans, Erik De Clercq, Christophe Pannecouque, Luis Menéndez-Arias, Peng Zhan, Xinyong Liu. 5-Hydroxypyrido[2,3-b]pyrazin-6(5H)-one derivatives as novel dual inhibitors of HIV-1 reverse transcriptase-associated ribonuclease H and integrase. European Journal of Medicinal Chemistry 2018, 155 , 714-724. https://doi.org/10.1016/j.ejmech.2018.06.036
- Cyrus M. Lacbay, Michael Menni, Jean A. Bernatchez, Matthias Götte, Youla S. Tsantrizos. Pharmacophore requirements for HIV-1 reverse transcriptase inhibitors that selectively “Freeze” the pre-translocated complex during the polymerization catalytic cycle. Bioorganic & Medicinal Chemistry 2018, 26
(8)
, 1713-1726. https://doi.org/10.1016/j.bmc.2018.02.017
- Yan Wang, Jing Tang, Zhengqiang Wang, Robert J. Geraghty. Metal-chelating 3-hydroxypyrimidine-2,4-diones inhibit human cytomegalovirus pUL89 endonuclease activity and virus replication. Antiviral Research 2018, 152 , 10-17. https://doi.org/10.1016/j.antiviral.2018.01.015
- Sonia de Castro, María-José Camarasa. Polypharmacology in HIV inhibition: can a drug with simultaneous action against two relevant targets be an alternative to combination therapy?. European Journal of Medicinal Chemistry 2018, 150 , 206-227. https://doi.org/10.1016/j.ejmech.2018.03.007
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Major chemotypes reported as HIV RNase H active site inhibitors. Chemotypes 4–7 reflect a pharmacophore model consisting of a chelating triad (magenta) and an aryl or biaryl moiety (cyan) connected through a methylene or amino linker.
Figure 2
Figure 2. Design of a novel HID scaffold 9 based on the pharmacophore model of 4–7.
Scheme 1
Scheme 1. Synthesis of C6 Arylmethyl HID Analogues 20a–taScheme aReagents and conditions: (a) NaBH4, THF, rt, 12 h, 87%; (b) n-BuLi, DMF, THF, −78 °C–rt, overnight, 76%; (c) TsNHNH2, toluene, 80 °C, 2 h; (d) boronic acid, K2CO3, 1,4-dioxane, 110 °C, 3–5 h, 40–55%; (e) PPTS, toluene, reflux, 6–12 h, 75–90%; (f) OsO4, NMO, t-BuOH/acetone/H2O, rt, 2–6 h, 60–81%; (g) NaIO4, RuCl3, CH3CN/CCl4/H2O, rt, 2–4 h, 67–85%; (h) ArB(OH)2, Pd(PPh3)4, K2CO3, EtOH/H2O, microwave, 120–150 °C, 20–30 min, 60–75%; (i) NH2OTHP, CDI, toluene, reflux, 12 h; (j) p-TSA hydrate, MeOH, 2–3 h, rt, 35–55% over two steps.
Figure 3
Figure 3. Binding mode of compound 20i. Left: structure of full length RT with two subunits p66 and p51 (orange). p66 subunit comprises fingers (blue), palm (red), thumb (green), connection (yellow), and RNase H (magenta) domains. Compounds 7 (green) and 20i (cyan) are docked into the RNase H active site. Right: a close-up view of RNase H active site with predicted binding mode of compound 7 (green) and 20i (cyan). Metal cofactors (Mn2+) are colored in gray, with the active site residues (D443, E478, D498 and D549) in yellow. Pictures were generated using PyMol. (41)
References
This article references 55 other publications.
- 1AIDSinfo; UNAIDS: Geneva 2013; http://www.unaids.org/en/dataanalysis/datatools/aidsinfo/.There is no corresponding record for this reference.
- 2Barouch, D. H. Challenges in the development of an HIV-1 vaccine Nature 2008, 455, 613– 619There is no corresponding record for this reference.
- 3Schiffner, T.; Sattentau, Q. J.; Dorrell, L. Development of prophylactic vaccines against HIV-1 Retrovirology 2013, 10, 72There is no corresponding record for this reference.
- 4Zhang, J.; Crumpacker, C. Eradication of HIV and Cure of AIDS, Now and How? Front. Immunol. 2013, 4, 3374Eradication of HIV and Cure of AIDS, Now and How?Zhang Jielin; Crumpacker ClydeFrontiers in immunology (2013), 4 (), 337 ISSN:1664-3224.Recent studies have highlighted the importance of eradication of human immunodeficiency virus (HIV) and cure of acquired immunodeficiency syndrome (AIDS). However, a pivotal point that the patient immunity controls HIV reactivation after highly active anti-retroviral therapy [HAART or combination anti-retroviral therapy (cART)] remains less well addressed. In spite of the fact that both innate and adaptive immunities are indispensable and numerous cells participate in the anti-HIV immunity, memory CD4 T-cells are indisputably the key cells organizing all immune actions against HIV while being the targets of HIV. Here we present a view and multidisciplinary approaches to HIV/AIDS eradication and cure. We aim at memory CD4 T-cells, utilizing the stem cell properties of these cells to reprogram an anti-HIV memory repertoire to eliminate the viral reservoir, toward achieving an AIDS-free world.
- 5Siliciano, J. D.; Siliciano, R. F. HIV-1 eradication strategies: design and assessment Curr. Opin. HIV AIDS 2013, 8, 318– 3255HIV-1 eradication strategies: design and assessmentSiliciano Janet D; Siliciano Robert FCurrent opinion in HIV and AIDS (2013), 8 (4), 318-25 ISSN:.PURPOSE OF REVIEW: Recent developments have generated renewed interest in the possibility of curing HIV-1 infection. This review describes some of the practical challenges that will need to be overcome if curative strategies are to be successful. RECENT FINDINGS: The latent reservoir for HIV-1 in resting memory CD4 T cells is the major barrier to curing the infection. The most widely discussed approach to curing the infection involves finding agents that reverse latency in resting CD4 T cells, with the assumption that the cells will then die from viral cytopathic effects or be lysed by host cytolytic T lymphocytes (CTLs). A major challenge is the development of in-vitro models that can be used to explore mechanisms and identify latency-reversing agents (LRAs). Although several models have been developed, including primary cell models, none of them may fully capture the quiescent state of the cells that harbour latent HIV-1 in vivo. An additional problem is that LRAs that do not cause T-cell activation may not lead to the death of infected cells. Finally, measuring the effects of LRAs in vivo is complicated by the lack of correlation between different assays for the latent reservoir. SUMMARY: Progress on these practical issues is essential to finding a cure.
- 6Xing, S.; Siliciano, R. F. Targeting HIV latency: pharmacologic strategies toward eradication Drug Discovery Today 2013, 18, 541– 551There is no corresponding record for this reference.
- 7Beilhartz, G. L.; Gotte, M. HIV-1 Ribonuclease H: Structure, Catalytic Mechanism and Inhibitors Viruses 2010, 2, 900– 9267HIV-1 ribonuclease H: structure, catalytic mechanism and inhibitorsBeilhartz, Greg L.; Gotte, MatthiasViruses (2010), 2 (), 900-926CODEN: VIRUBR; ISSN:1999-4915. (Molecular Diversity Preservation International)A review. Since the human immunodeficiency virus (HIV) was discovered as the etiol. agent of acquired immunodeficiency syndrome (AIDS), it has encouraged much research into antiviral compds. The reverse transcriptase (RT) of HIV has been a main target for antiviral drugs. However, all drugs developed so far inhibit the polymerase function of the enzyme, while none of the approved antiviral agents inhibit specifically the necessary RNase H (RNase H) function of RT. This review provides a background on structure-function relationships of HIV-1 RNase H, as well as an outline of current attempts to develop novel, potent chemotherapeutics against a difficult drug target.
- 8Sarafianos, S. G.; Marchand, B.; Das, K.; Himmel, D. M.; Parniak, M. A.; Hughes, S. H.; Arnold, E. Structure and function of HIV-1 reverse transcriptase: molecular mechanisms of polymerization and inhibition J. Mol. Biol. 2009, 385, 693– 7138Structure and function of HIV-1 reverse transcriptase: Molecular mechanisms of polymerization and inhibitionSarafianos, Stefan G.; Marchand, Bruno; Das, Kalyan; Himmel, Daniel M.; Parniak, Michael A.; Hughes, Stephen H.; Arnold, EddyJournal of Molecular Biology (2009), 385 (3), 693-713CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. The rapid replication of the HIV-1 virus and errors made during viral replication cause the virus to evolve rapidly in patients, making the problems of vaccine development and drug therapy particularly challenging. In the absence of an effective vaccine, drugs are the only useful treatment. Anti-HIV drugs work; so far drug therapy has saved more than 3 × 106 yr of life. Unfortunately, HIV-1 virus develops resistance to all of the available drugs. Although a no. of useful anti-HIV drugs have been approved for use in patients, the problems assocd. with drug toxicity and the development of resistance means that the search for new drugs is an ongoing process. The 3 viral enzymes, reverse transcriptase (RT), integrase (IN), and protease (PR) are all good drug targets. Two distinct types of RT inhibitors, both of which block the polymerase activity of RT, have been approved to treat HIV-1 infections, nucleoside analogs (NRTIs) and non-nucleosides (NNRTIs), and there are promising leads for compds. that either block the RNase H activity or block the polymerase in other ways. A better understanding of the structure and function(s) of RT and of the mechanism(s) of inhibition can be used to generate better drugs; in particular, drugs that are effective against the current drug-resistant strains of HIV-1.
- 9Cihlar, T.; Ray, A. S. Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudine Antiviral Res. 2010, 85, 39– 589Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudineCihlar, Tomas; Ray, Adrian S.Antiviral Research (2010), 85 (1), 39-58CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. Twenty-five years ago, nucleoside analog 3'-azidothymidine (AZT) was shown to efficiently block the replication of HIV in cell culture. Subsequent studies demonstrated that AZT acts via the selective inhibition of HIV reverse transcriptase (RT) by its triphosphate metabolite. These discoveries have established the first class of antiretroviral agents: nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs). Over the years that followed, NRTIs evolved into the main component of antiretroviral drug combinations that are now used for the treatment of all populations of HIV infected patients. A total of thirteen NRTI drug products are now available for clin. application: eight individual NRTIs, four fixed-dose combinations of two or three NRTIs, and one complete fixed-dose regimen contg. two NRTIs and one non-nucleoside RT inhibitor. Multiple NRTIs or their prodrugs are in various stages of clin. development and new potent NRTIs are still being identified through drug discovery efforts. This article will review basic principles of the in vitro and in vivo pharmacol. of NRTIs, discuss their clin. use including limitations assocd. with long-term NRTI therapy, and describe newly identified NRTIs with promising pharmacol. profiles highlighting those in the development pipeline. This article forms part of a special issue of Antiviral Research marking the 25th anniversary of antiretroviral drug discovery and development, vol. 85, issue 1, 2010.
- 10de Bethune, M. P. Non-nucleoside reverse transcriptase inhibitors (NNRTIs), their discovery, development, and use in the treatment of HIV-1 infection: a review of the last 20 years (1989–2009) Antiviral Res. 2010, 85, 75– 90There is no corresponding record for this reference.
- 11Corona, A.; Masaoka, T.; Tocco, G.; Tramontano, E.; Le Grice, S. F. Active site and allosteric inhibitors of the ribonuclease H activity of HIV reverse transcriptase Future Med. Chem. 2013, 5, 2127– 213911Active site and allosteric inhibitors of the ribonuclease H activity of HIV reverse transcriptaseCorona, Angela; Masaoka, Takashi; Tocco, Graziella; Tramontano, Enzo; Le Grice, Stuart FJFuture Medicinal Chemistry (2013), 5 (18), 2127-2139CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. Despite the wealth of information available for the reverse transcriptase (RT)-assocd. RNase H (RNaseH) domain of lentiviruses, gamma-retroviruses and long terminal repeat contg. retrotransposons, exploiting this information in the form of an RNaseH inhibitor with high specificity and low cellular toxicity has been disappointing. However, it is now becoming increasingly evident that the two-subunit HIV-1 RT is a highly versatile enzyme, undergoing major structural alterations in order to interact with, position and ultimately hydrolyze the RNA component of an RNA/DNA hybrid. Thus, in addn. to targeting the RNaseH active site, identifying small mols. that bind elsewhere and disrupt catalysis allosterically by impairing conformational flexibility is gaining increased attention. This review summarizes current progress towards development of both active site and allosteric RNaseH inhibitors.
- 12Ilina, T.; Labarge, K.; Sarafianos, S. G.; Ishima, R.; Parniak, M. A. Inhibitors of HIV-1 Reverse Transcriptase-Associated Ribonuclease H Activity Biology (Basel, Switzerland) 2012, 1, 521– 54112Inhibitors of HIV-1 reverse transcriptase - associated ribonuclease H activityIlina, Tatiana; LaBarge, Krystal; Sarafianos, Stefan G.; Ishima, Rieko; Parniak, Michael A.Biology (Basel, Switzerland) (2012), 1 (3), 521-541CODEN: BBSIBX; ISSN:2079-7737. (MDPI AG)A review. HIV-1 enzyme reverse transcriptase (RT) is a major target for antiviral drug development, with over half of current FDA-approved therapeutics against HIV infection targeting the DNA polymerase activity of this enzyme. HIV-1 RT is a multifunctional enzyme that has RNA and DNA dependent polymerase activity, along with RNase H (RNase H) activity. The latter is responsible for degrdn. of the viral genomic RNA template during first strand DNA synthesis to allow completion of reverse transcription and the viral dsDNA. While the RNase H activity of RT has been shown to be essential for virus infectivity, all currently used drugs directed at RT inhibit the polymerase activity of the enzyme; none target RNase H. In the last decade, the increasing prevalence of HIV variants resistant to clin. used antiretrovirals has stimulated the search for inhibitors directed at stages of HIV replication different than those targeted by current drugs. HIV RNase H is one such novel target and, over the past few years, significant progress has been made in identifying and characterizing new RNase H inhibitor pharmacophores. In this review we focus mainly on the most potent low micromolar potency compds., as these provide logical bases for further development. We also discuss why HIV RNase H has been a difficult target for antiretroviral drug development.
- 13Klumpp, K.; Hang, J. Q.; Rajendran, S.; Yang, Y. L.; Derosier, A.; In, P. W. K.; Overton, H.; Parkes, K. E. B.; Cammack, N.; Martin, J. A. Two-metal ion mechanism of RNA cleavage by HIV RNase H and mechanism-based design of selective HIV RNase H inhibitors Nucleic Acids Res. 2003, 31, 6852– 685913Two-metal ion mechanism of RNA cleavage by HIV RNase H and mechanism-based design of selective HIV RNase H inhibitorsKlumpp, Klaus; Hang, Julie Qi; Rajendran, Surendran; Yang, Yanli; Derosier, Andre; In, Philippe Wong Kai; Overton, Hilary; Parkes, Kevin E. B.; Cammack, Nick; Martin, Joseph A.Nucleic Acids Research (2003), 31 (23), 6852-6859CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Human immunodeficiency virus (HIV) RNase H activity is essential for the synthesis of viral DNA by HIV reverse transcriptase (HIV-RT). RNA cleavage by RNase H requires the presence of divalent metal ions, but the role of metal ions in the mechanism of RNA cleavage has not been resolved. We measured HIV RNase H activity assocd. with HIV-RT protein in the presence of different concns. of either Mg2+, Mn2+, Co2+ or a combination of these divalent metal ions. Polymerase-independent HIV RNase H was similar to or more active with Mn2+ and Co2+ compared with Mg2+. Activation of RNase H by these metal ions followed sigmoidal dose-response curves suggesting cooperative metal ion binding. Titrn. of Mg2+-bound HIV RNase H with Mn2+ or Co2+ ions generated bell-shaped activity dose-response curves. Higher activity could be achieved through simultaneous binding of more than one divalent metal ion at intermediate Mn2+ and Co2+ concns., and complete replacement of Mg2+ occurred at higher Mn2+ or Co2+ concns. These results are consistent with a two-metal ion mechanism of RNA cleavage as previously suggested for a no. of polymerase-assocd. nucleases. In contrast, the structurally highly homologous RNase HI from Escherichia coli is most strongly activated by Mg2+, is significantly inhibited by submillimolar concns. of Mn2+ and most probably cleaves RNA via a one-metal ion mechanism. Based on this difference in active site structure, a series of small mol. N-hydroxyimides was identified with significant enzyme inhibitory potency and selectivity for HIV RNase H.
- 14Himmel, D. M.; Sarafianos, S. G.; Dharmasena, S.; Hossain, M. M.; McCoy-Simandle, K.; Ilina, T.; Clark, A. D.; Knight, J. L.; Julias, J. G.; Clark, P. K.; Krogh-Jespersen, K.; Levy, R. M.; Hughes, S. H.; Parniak, M. A.; Arnold, E. HIV-1 reverse transcriptase structure with RNase H inhibitor dihydroxy benzoyl naphthyl hydrazone bound at a novel site ACS Chem. Biol. 2006, 1, 702– 71214HIV-1 Reverse Transcriptase Structure with RNase H Inhibitor Dihydroxy Benzoyl Naphthyl Hydrazone Bound at a Novel SiteHimmel, Daniel M.; Sarafianos, Stefan G.; Dharmasena, Sanjeewa; Hossain, Mohammed M.; McCoy-Simandle, Kessler; Ilina, Tatiana; Clark, Arthur D., Jr.; Knight, Jennifer L.; Julias, John G.; Clark, Patrick K.; Krogh-Jespersen, Karsten; Levy, Ronald M.; Hughes, Stephen H.; Parniak, Michael A.; Arnold, EddyACS Chemical Biology (2006), 1 (11), 702-712CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The rapid emergence of drug-resistant variants of human immunodeficiency virus, type 1 (HIV-1), has limited the efficacy of anti-acquired immune deficiency syndrome (AIDS) treatments, and new lead compds. that target novel binding sites are needed. The authors have detd. the 3.15 Å resoln. crystal structure of HIV-1 reverse transcriptase (RT) complexed with dihydroxy benzoyl naphthyl hydrazone (DHBNH), an HIV-1 RT RNase H (RNH) inhibitor (RNHI). DHBNH is effective against a variety of drug-resistant HIV-1 RT mutants. While DHBNH has little effect on most aspects of RT-catalyzed DNA synthesis, at relatively high concns. it does inhibit the initiation of RNA-primed DNA synthesis. Although primarily an RNHI, DHBNH binds >50 Å away from the RNH active site, at a novel site near both the polymerase active site and the nonnucleoside RT inhibitor (NNRTI) binding pocket. When DHBNH binds, both Tyr181 and Tyr188 remain in the conformations seen in unliganded HIV-1 RT. DHBNH interacts with conserved residues (Asp186, Trp229) and has substantial interactions with the backbones of several less well-conserved residues. On the basis of this structure, the authors designed substituted DHBNH derivs. that interact with the NNRTI-binding pocket. These compds. inhibit both the polymerase and RNH activities of RT.
- 15Fuji, H.; Urano, E.; Futahashi, Y.; Hamatake, M.; Tatsumi, J.; Hoshino, T.; Morikawa, Y.; Yamamoto, N.; Komano, J. Derivatives of 5-nitro-furan-2-carboxylic acid carbamoylmethyl ester inhibit RNase H activity associated with HIV-1 reverse transcriptase J. Med. Chem. 2009, 52, 1380– 1387There is no corresponding record for this reference.
- 16Tramontano, E.; Esposito, F.; Badas, R.; Di Santo, R.; Costi, R.; La Colla, P. 6-[1-(4-Fluorophenyl)methyl-1H-pyrrol-2-yl)]-2,4-dioxo-5-hexenoic acid ethyl ester a novel diketo acid derivative which selectively inhibits the HIV-1 viral replication in cell culture and the ribonuclease H activity in vitro Antiviral Res. 2005, 65, 117– 124166-[1-(4-Fluorophenyl)methyl-1H-pyrrol-2-yl]-2,4-dioxo-5-hexenoic acid ethyl ester a novel diketo acid derivative which selectively inhibits the HIV-1 viral replication in cell culture and the ribonuclease H activity in vitroTramontano, Enzo; Esposito, Francesca; Badas, Roberta; Di Santo, Roberto; Costi, Roberta; La Colla, PaoloAntiviral Research (2005), 65 (2), 117-124CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)The human immunodeficiency virus-type 1 (HIV-1) reverse transcriptase (RT) is a multifunctional enzyme which displays DNA polymerase activity, which recognizes RNA and DNA templates, and a degradative RNase H activity. While both RT functions are required for retroviral replication, until now only the polymerase function has been widely explored as drug target. We have identified a novel diketo acid deriv., 6-[1-(4-fluorophenyl)methyl-1H-pyrrol-2-yl]-2,4-dioxo-5-hexenoic acid Et ester (RDS 1643), which inhibits in enzyme assays the HIV-1 RT-assocd. polymerase-independent RNase H activity but has no effect on the HIV-1 RT-assocd. RNA-dependent DNA polymerase (RDDP) activity and on the RNase H activities displayed by the Avian Myeloblastosis Virus and E. coli. Time-dependence studies revealed that the compd. is active independently on the order of its addn. to the reaction mixt., and inhibition kinetics studies demonstrated that RDS 1643 inhibits the RNase H activity noncompetitively, with a KI value of 17 μM. When RDS 1643 was combined with non-nucleoside RT inhibitors (NNRTI), such as efavirenz and nevirapine, results indicated that RDS 1643 does not affect the NNRTIs anti-RDDP activity and that, vice versa, the NNRTIs do not alter the RNase H inhibition by RDS 1643. When assayed on the viral replication in cell-based assays, RDS 1643 inhibited the HIV-1IIIB strain with an EC50 of 14 μM. Similar results were obtained against the Y181C and Y181C/K103N HIV-1 NNRTI resistant mutant strains. RDS 1643 may be the first HIV-1 inhibitor selectively targeted to the viral RT-assocd. RNase-H function.
- 17Lansdon, E. B.; Liu, Q.; Leavitt, S. A.; Balakrishnan, M.; Perry, J. K.; Lancaster-Moyer, C.; Kutty, N.; Liu, X.; Squires, N. H.; Watkins, W. J.; Kirschberg, T. A. Structural and binding analysis of pyrimidinol carboxylic acid and N-hydroxy quinazolinedione HIV-1 RNase H inhibitors Antimicrob. Agents Chemother. 2011, 55, 2905– 2915There is no corresponding record for this reference.
- 18Williams, P. D.; Staas, D. D.; Venkatraman, S.; Loughran, H. M.; Ruzek, R. D.; Booth, T. M.; Lyle, T. A.; Wai, J. S.; Vacca, J. P.; Feuston, B. P.; Ecto, L. T.; Flynn, J. A.; DiStefano, D. J.; Hazuda, D. J.; Bahnck, C. M.; Himmelberger, A. L.; Dornadula, G.; Hrin, R. C.; Stillmock, K. A.; Witmer, M. V.; Miller, M. D.; Grobler, J. A. Potent and selective HIV-1 ribonuclease H inhibitors based on a 1-hydroxy-1,8-naphthyridin-2(1H)-one scaffold Bioorg. Med. Chem. Lett. 2010, 20, 6754– 6757There is no corresponding record for this reference.
- 19Gerondelis, P.; Johns, B. A., The development of novel pyrido-pyrimidinone antiretrovirals with selective activity against HIV ribonuclease H. , Cold Spring Harbor Laboratories Conference on Retroviruses, Cold Spring Harbor, NY, May 21–26, 2012; Cold Spring Harbor Press: Cold Spring Harbor, NY, 2012.There is no corresponding record for this reference.
- 20Beilhartz, G. L.; Ngure, M.; Johns, B. A.; DeAnda, F.; Gerondelis, P.; Gotte, M. Inhibition of the Ribonuclease H Activity of HIV-1 Reverse Transcriptase by GSK5750 Correlates with Slow Enzyme–Inhibitor Dissociation J. Biol. Chem. 2014, 289, 16270– 16277There is no corresponding record for this reference.
- 21Chen, Y. L.; Tang, J.; Kesler, M. J.; Sham, Y. Y.; Vince, R.; Geraghty, R. J.; Wang, Z. Q. The design, synthesis and biological evaluations of C-6 or C-7 substituted 2-hydroxyisoquinoline-1,3-diones as inhibitors of hepatitis C virus Bioorg. Med. Chem. 2012, 20, 467– 479There is no corresponding record for this reference.
- 22Billamboz, M.; Bailly, F.; Lion, C.; Calmels, C.; Andreola, M. L.; Witvrouw, M.; Christ, F.; Debyser, Z.; De Luca, L.; Chimirri, A.; Cotelle, P. 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as inhibitors of HIV-1 integrase and reverse transcriptase RNase H domain: influence of the alkylation of position 4 Eur. J. Med. Chem. 2011, 46, 535– 546There is no corresponding record for this reference.
- 23Billamboz, M.; Suchaud, V.; Bailly, F.; Lion, C.; Demeulemeester, J.; Calmels, C.; Andreola, M. L.; Christ, F.; Debyser, Z.; Cotelle, P. 4-Substituted 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as a Novel Class of HIV-1 Integrase Inhibitors ACS Med. Chem. Lett. 2013, 4, 41– 46There is no corresponding record for this reference.
- 24Desimmie, B. A.; Demeulemeester, J.; Suchaud, V.; Taltynov, O.; Billamboz, M.; Lion, C.; Bailly, F.; Strelkov, S. V.; Debyser, Z.; Cotelle, P.; Christ, F. 2-Hydroxyisoquinoline-1,3(2H,4H)-diones (HIDs), Novel Inhibitors of HIV Integrase with a High Barrier to Resistance ACS Chem. Biol. 2013, 8, 1187– 1194There is no corresponding record for this reference.
- 25Suchaud, V.; Bailly, F.; Lion, C.; Calmels, C.; Andréola, M.-L.; Christ, F.; Debyser, Z.; Cotelle, P. Investigation of a Novel Series of 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as Human Immunodeficiency Virus Type 1 Integrase Inhibitors J. Med. Chem. 2014, 57, 4640– 4660http://dx.doi.org/10.1021/jm500109zThere is no corresponding record for this reference.
- 26Billamboz, M.; Bailly, F.; Lion, C.; Touati, N.; Vezin, H.; Calmels, C.; Andreola, M. L.; Christ, F.; Debyser, Z.; Cotelle, P. Magnesium Chelating 2-Hydroxyisoquinoline-1,3(2H,4H)-diones, as Inhibitors of HIV-1 Integrase and/or the HIV-1 Reverse Transcriptase Ribonuclease H Domain: Discovery of a Novel Selective Inhibitor of the Ribonuclease H Function J. Med. Chem. 2011, 54, 1812– 182426Magnesium Chelating 2-Hydroxyisoquinoline-1,3(2H,4H)-diones, as Inhibitors of HIV-1 Integrase and/or the HIV-1 Reverse Transcriptase Ribonuclease H Domain: Discovery of a Novel Selective Inhibitor of the Ribonuclease H FunctionBillamboz, Muriel; Bailly, Fabrice; Lion, Cedric; Touati, Nadia; Vezin, Herve; Calmels, Christina; Andreola, Marie-Line; Christ, Frauke; Debyser, Zeger; Cotelle, PhilippeJournal of Medicinal Chemistry (2011), 54 (6), 1812-1824CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)2-Hydroxyisoquinoline-1,3(2H,4H)-dione was recently discovered as a scaffold for the inhibition of HIV-1 integrase and the RNase H function of HIV-1 reverse transcriptase. First, we investigate its interaction with Mg2+ and Mn2+ using different spectroscopic techniques and report that 2-hydroxyisoquinoline-1,3(2H,4H)-dione forms a 1:1 complex with Mg2+ but a 1:2 complex with Mn2+. The complex formation requires enolization of the ligand. ESR spectroscopy shows a redox reaction between the ligand and Mn2+ producing superoxide anions. Second, 2-hydroxyisoquinoline-1,3(2H,4H)-dione, its magnesium complex, and its 4-Me and 2-hydroxy-4-methoxycarbonylisoquinoline-1,3(2H,4H)-diones were tested as inhibitors of HIV-1 integrase, reverse transcriptase RNase H, and DNA polymerase functions. Their antiviral activities were evaluated and 2-hydroxy-4-methoxycarbonyl-isoquinoline-1,3(2H,4H)-dione was found to inhibit the viral replication of HIV-1 in MT-4 cells. Cross-resistance was measured for this compd. on three different viral strains. Exptl. data suggest that the antiviral activity of 2-hydroxy-4-methoxycarbonylisoquinoline-1,3(2H,4H)-dione is probably due to the RNase H inhibition.
- 27Billamboz, M.; Bailly, F.; Barreca, M. L.; De Luca, L.; Mouscadet, J. F.; Calmels, C.; Andreola, M. L.; Witvrouw, M.; Christ, F.; Debyser, Z.; Cotelle, P. Design, Synthesis, and Biological Evaluation of a Series of 2-Hydroxyisoquinoline-1,3(2H,4H)-diones as Dual Inhibitors of Human Immunodeficiency Virus Type 1 Integrase and the Reverse Transcriptase RNase H Domain J. Med. Chem. 2008, 51, 7717– 7730There is no corresponding record for this reference.
- 28Barluenga, J.; Tomas-Gamasa, M.; Aznar, F.; Valdes, C. Metal-free carbon–carbon bond-forming reductive coupling between boronic acids and tosylhydrazones Nature Chem. 2009, 1, 494– 49928Metal-free carbon-carbon bond-forming reductive coupling between boronic acids and tosylhydrazonesBarluenga, Jose; Tomas-Gamasa, Maria; Aznar, Fernando; Valdes, CarlosNature Chemistry (2009), 1 (6), 494-499, S494/1-S494/46CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The formation of carbon-carbon bonds is a fundamental transformation in org. synthesis. In spite of the myriad methods available, advantageous methodologies in terms of selectivity, availability of starting materials, operational simplicity, functional-group tolerance, environmental sustainability and economy are in const. demand. In this context, the development of new cross-coupling reactions that use catalysts based on inexpensive and non-toxic metals is attracting increasing attention. Similarly, efficient processes that do not require a metal catalyst are of extraordinary interest. Here, we report a new and efficient metal-free carbon-carbon bond-forming coupling between tosylhydrazones and boronic acids. This reaction is very general and functional-group tolerant. As the required tosylhydrazones are easily generated from carbonyl compds., it can be seen as a reductive coupling of carbonyls, a process of high synthetic relevance that requires several steps using other methodologies.
- 29Vanrheenen, V.; Cha, D. Y.; Hartley, W. M. Catalytic Osmium-Tetroxide Oxidation of Olefins—cis-1,2-Cyclohexanediol Org. Synth. 1988, 50–9, 342– 348There is no corresponding record for this reference.
- 30Vanrheenen, V.; Kelly, R. C.; Cha, D. Y. Improved Catalytic OsO4 Oxidation of Olefins to cis-1,2-Glycols Using Tertiary Amine Oxides as Oxidant Tetrahedron Lett. 1976, 1973– 1976There is no corresponding record for this reference.
- 31Yang, D.; Zhang, C. Ruthenium-catalyzed oxidative cleavage of olefins to aldehydes J. Org. Chem. 2001, 66, 4814– 481831Ruthenium-Catalyzed Oxidative Cleavage of Olefins to AldehydesYang, Dan; Zhang, ChiJournal of Organic Chemistry (2001), 66 (14), 4814-4818CODEN: JOCEAH; ISSN:0022-3263. (American Chemical Society)Three oxidn. protocols have been developed to cleave olefins to carbonyl compds. with ruthenium trichloride as catalyst (3.5 mol %). These methods convert olefins that are not fully substituted to aldehydes rather than carboxylic acids. While aryl olefins were cleaved to arom. aldehydes in excellent yields by using the system of RuCl3-Oxone-NaHCO3 in CH3CN-H2O (1.5:1), aliph. olefins were converted into alkyl aldehydes with RuCl3-NaIO4 in 1,2-dichloroethane-H2O (1:1) in good to excellent yields. It is noteworthy that terminal aliph. olefins were cleaved to the corresponding aldehydes in excellent yields by using RuCl3-NaIO4 in CH3CN-H2O (6:1).
- 32Parniak, M. A.; Min, K. L.; Budihas, S. R.; Le Grice, S. F.; Beutler, J. A. A fluorescence-based high-throughput screening assay for inhibitors of human immunodeficiency virus-1 reverse transcriptase-associated ribonuclease H activity Anal. Biochem. 2003, 322, 33– 39There is no corresponding record for this reference.
- 33Parniak, M. A.; Min, K. L. Substrate for assaying ribonuclease H activity. US 7,186,520 March6, 2007.There is no corresponding record for this reference.
- 34Parniak, M. A.; Min, K. L. Method of identifying or characterizing a compound that modulates ribonuclease H activity. US 7,439,035 October21, 2008.There is no corresponding record for this reference.
- 35Nowotny, M. Retroviral integrase superfamily: the structural perspective EMBO Rep. 2009, 10, 144– 15135Retroviral integrase superfamily: the structural perspectiveNowotny, MarcinEMBO Reports (2009), 10 (2), 144-151CODEN: ERMEAX; ISSN:1469-221X. (Nature Publishing Group)A review. The retroviral integrase superfamily (RISF) comprises numerous important nucleic acid-processing enzymes, including transposases, integrases and various nucleases. These enzymes are involved in a wide range of processes such as transposition, replication and repair of DNA, homologous recombination, and RNA-mediated gene silencing. Two out of the four enzymes that are encoded by the human immunodeficiency virus, RNase H1 and integrase, are members of this superfamily. RISF enzymes act on various substrates, and yet show remarkable mechanistic and structural similarities. All share a common fold of the catalytic core and the active site, which is composed primarily of carboxylate residues. Here, RISF proteins are presented from a structural perspective, describing the individual members and the common and divergent elements of their structures, as well as the mechanistic insights gained from the structures of RNase H1 enzyme complexes with RNA/DNA hybrids.
- 36Barreca, M. L.; Ferro, S.; Rao, A.; De Luca, L.; Zappala, M.; Monforte, A. M.; Debyser, Z.; Witvrouw, M.; Chimirri, A. Pharmacophore-based design of HIV-1 integrase strand-transfer inhibitors J. Med. Chem. 2005, 48, 7084– 7088There is no corresponding record for this reference.
- 37Abram, M. E.; Parniak, M. A. Virion instability of human immunodeficiency virus type 1 reverse transcriptase (RT) mutated in the protease cleavage site between RT p51 and the RT RNase H domain J. Virol. 2005, 79, 11952– 1196137Virion instability of human immunodeficiency virus type 1 reverse transcriptase (RT) mutated in the protease cleavage site between RT p51 and the RT RNase H domainAbram, Michael E.; Parniak, Michael A.Journal of Virology (2005), 79 (18), 11952-11961CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Each of the human immunodeficiency virus type 1 (HIV-1) pol-encoded enzymes, protease (PR), reverse transcriptase (RT), and integrase (IN), is active only as a dimer (or higher-order oligomer in the case of IN), but only RT comprises subunits of different masses. RT is a heterodimer of 66-kDa and 51-kDa subunits. The latter is formed by HIV PR-catalyzed cleavage of p66 during virion maturation, resulting in the removal of the RNase H (RNH) domain of a p66 subunit. In order to study the apparent need for RT heterodimers in the context of the virion, we introduced a variety of mutations in the RT p51-RNH protease cleavage site of an infectious HIV-1 mol. clone. Surprisingly, rather than leading to virions with increased RT p66 content, most of the mutations resulted in significantly attenuated virus that contained greatly decreased levels of RT that in many cases was primarily p51 RT. IN levels were also reduced in several mutants. However, most mutants showed normal levels of the Pr160gag-pol precursor polyprotein, suggesting that reduced virion RT arose from proteolytic instability rather than decreased incorporation. Mutant virion p24 Gag levels were equiv. to wild type, indicating that Gag incorporation and processing were not affected. Repeated passage of MT-2 cells exposed to mutant viruses led to the appearance of virus with improved replication capacity; these virions contained normally processed RT at near-wild-type levels. These results imply that addnl. proteolytic processing of RT to the p66/p51 heterodimer is essential to provide proteolytic stability of RT during HIV-1 maturation.
- 38Parikh, U. M.; Koontz, D. L.; Chu, C. K.; Schinazi, R. F.; Mellors, J. W. In vitro activity of structurally diverse nucleoside analogs against human immunodeficiency virus type 1 with the K65R mutation in reverse transcriptase Antimicrob. Agents Chemother. 2005, 49, 1139– 1144There is no corresponding record for this reference.
- 39Su, H.-P.; Yan, Y.; Prasad, G. S.; Smith, R. F.; Daniels, C. L.; Abeywickrema, P. D.; Reid, J. C.; Loughran, H. M.; Kornienko, M.; Sharma, S.; Grobler, J. A.; Xu, B.; Sardana, V.; Allison, T. J.; Williams, P. D.; Darke, P. L.; Hazuda, D. J.; Munshi, S. Structural Basis for the Inhibition of RNase H Activity of HIV-1 Reverse Transcriptase by RNase H Active Site-Directed Inhibitors J. Virol. 2010, 84, 7625– 763339Structural basis for the inhibition of RNase H activity of HIV-1 reverse transcriptase by RNase H active site-directed inhibitorsSu, Hua-Poo; Yan, Youwei; Prasad, G. Sridhar; Smith, Robert F.; Daniels, Christopher L.; Abeywickrema, Pravien D.; Reid, John C.; Loughran, H. Marie; Kornienko, Maria; Sharma, Sujata; Grobler, Jay A.; Xu, Bei; Sardana, Vinod; Allison, Timothy J.; Williams, Peter D.; Darke, Paul L.; Hazuda, Daria J.; Munshi, SanjeevJournal of Virology (2010), 84 (15), 7625-7633CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)HIV/AIDS continues to be a menace to public health. Several drugs currently on the market have successfully improved the ability to manage the viral burden in infected patients. However, new drugs are needed to combat the rapid emergence of mutated forms of the virus that are resistant to existing therapies. Currently, approved drugs target three of the four major enzyme activities encoded by the virus that are crit. to the HIV life cycle. Although a no. of inhibitors of HIV RNase H activity have been reported, few inhibit by directly engaging the RNase H active site. Here, we describe structures of naphthyridinone-contg. inhibitors bound to the RNase H active site. This class of compds. binds to the active site via two metal ions that are coordinated by catalytic site residues, D443, E478, D498, and D549. The directionality of the naphthyridinone pharmacophore is restricted by the ordering of D549 and H539 in the RNase H domain. In addn., one of the naphthyridinone-based compds. was found to bind at a second site close to the polymerase active site and non-nucleoside/nucleotide inhibitor sites in a metal-independent manner. Further characterization, using fluorescence-based thermal denaturation and a crystal structure of the isolated RNase H domain reveals that this compd. can also bind the RNase H site and retains the metal-dependent binding mode of this class of mols. These structures provide a means for structurally guided design of novel RNase H inhibitors.
- 40Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes J. Med. Chem. 2006, 49, 6177– 619640Extra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 41PyMOL, The PyMOL Molecular Graphics System, version 1.5.0.4; Schrödinger, LLC: New York 2013.There is no corresponding record for this reference.
- 42Fletcher, R. S.; Holleschak, G.; Nagy, E.; Arion, D.; Borkow, G.; Gu, Z.; Wainberg, M. A.; Parniak, M. A. Single-step purification of recombinant wild-type and mutant HIV-1 reverse transcriptase Protein Express. Purif. 1996, 7, 27– 3242Single-step purification of recombinant wild-type and mutant HIV-1 reverse transcriptaseFletcher, Ronald S.; Holleschak, Gyula; Nagy, Eva; Arion, Dominique; Borkow, Gadi; Gu, Zhengxian; Wainberg, Mark A.; Parniak, Michael A.Protein Expression and Purification (1996), 7 (1), 27-32CODEN: PEXPEJ; ISSN:1046-5928. (Academic)A single-step method that enables the purifn. of HIV-1 recombinant reverse transcriptase (RT) directly from bacterial lysates in <2 h is described. Clarified lysates were applied to com. Q- and S-matrix cartridge columns connected in series. The columns were washed with low-salt buffer to remove unbound protein, then the Q column was removed and RT was eluted from the S column using a salt gradient. The purifn. was carried out with both medium-pressure and high-pressure chromatog. systems. The purifns. were carried out at room temp. near neutral pH, providing enzyme with high DNA polymerase specific activity. A crucial aspect of the procedure was the use of Tris buffer, a buffer that is normally incompatible in cation-exchange methods. The method was applicable for the purifn. of the p51/p66 heterodimer and the p51 and p66 homodimer forms of RT. This method was used to purify wild-type RT and several recombinant RT proteins contg. mutations correlated with dideoxynucleoside drug resistance.
- 43Gong, Q.; Menon, L.; Ilina, T.; Miller, L. G.; Ahn, J.; Parniak, M. A.; Ishima, R. Interaction of HIV-1 reverse transcriptase ribonuclease H with an acylhydrazone inhibitor Chem. Biol. Drug Des. 2011, 77, 39– 4743Interaction of HIV-1 reverse transcriptase ribonuclease H with an acylhydrazone inhibitorGong, Qingguo; Menon, Lakshmi; Ilina, Tatiana; Miller, Lena G.; Ahn, Jinwoo; Parniak, Michael A.; Ishima, RiekoChemical Biology & Drug Design (2010), 77 (1), 39-47CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)HIV-1 reverse transcriptase is a bifunctional enzyme, having both DNA polymerase (RNA- and DNA-dependent) and RNase H activities. HIV-1 reverse transcriptase has been an exceptionally important target for antiretroviral therapeutic development, and nearly half of the current clin. used antiretrovirals target reverse transcriptase DNA polymerase. However, no inhibitors of reverse transcriptase RNase H are on the market or in preclin. development. Several drug-like small mol. inhibitors of reverse transcriptase RNase H have been described, but little structural information is available about the interactions between reverse transcriptase RNase H and inhibitors that exhibit antiviral activity. In this report, we describe NMR studies of the interaction of a new RNase H inhibitor, BHMP07, with a catalytically active HIV-1 reverse transcriptase RNase H domain fragment. We carried out soln. NMR expts. to identify the interaction interface of BHMP07 with the RNase H domain fragment. Chem. shift changes of backbone amide signals at different BHMP07 concns. clearly demonstrate that BHMP07 mainly recognizes the substrate handle region in the RNase H fragment. Using RNase H inhibition assays and reverse transcriptase mutants, the binding specificity of BHMP07 was compared with another inhibitor, dihydroxy benzoyl naphthyl hydrazone. Our results provide a structural characterization of the RNase H inhibitor interaction and are likely to be useful for further improvements of the inhibitors.
- 44Dabora, J. M.; Marqusee, S. Equilibrium unfolding of Escherichia coli ribonuclease H: characterization of a partially folded state Protein Sci. 1994, 3, 1401– 140844Equilibrium unfolding of Escherichia coli ribonuclease H: characterization of a partially folded stateDabora, Jonathan M.; Marqusee, SusanProtein Science (1994), 3 (9), 1401-8CODEN: PRCIEI; ISSN:0961-8368.The equil. unfolding of Escherichia coli RNase HI (RNase H), a member of a family of enzymes the cleaves RNA from RNA:DNA hybrids, was examd. A completely synthetic gene was constructed that expresses a variant of the wild-type sequence with all 3 cysteines replaced with alanine. The resulting recombinant protein is active and folds reversibly. Denaturation studies monitored by CD and tryptophan fluorescence yield coincident curves that suggest that the equil. unfolding reaction is a 2-state process. Acid denaturation, however, reveals a cooperative transition at pH ∼1.8 to a partially folded state. This acid state can be further denatured in a reversible manner by the addn. of heat or urea as monitored by either CD or tryptophan fluorescence. Anal. ultracentrifugation studies indicate that the acid state of RNase H is both compact and monomeric. Although compact, the acid state does not resemble the native protein: the acid state displays a near-UV CD spectrum similar to the unfolded state and binds to and enhances the fluorescence of the dye 1-anilinonaphthalene 8-sulfonate much more than either the native or unfolded states. Therefore, the acid state of E. coli RNase H has the characteristics of a molten globule: it retains a high degree of secondary structure, remains compact, yet does not appear to contain a tightly packed core.
- 45Ndongwe, T. P.; Adedeji, A. O.; Michailidis, E.; Ong, Y. T.; Hachiya, A.; Marchand, B.; Ryan, E. M.; Rai, D. K.; Kirby, K. A.; Whatley, A. S.; Burke, D. H.; Johnson, M.; Ding, S.; Zheng, Y. M.; Liu, S. L.; Kodama, E.; Delviks-Frankenberry, K. A.; Pathak, V. K.; Mitsuya, H.; Parniak, M. A.; Singh, K.; Sarafianos, S. G. Biochemical, inhibition and inhibitor resistance studies of xenotropic murine leukemia virus-related virus reverse transcriptase Nucleic Acids Res. 2012, 40, 345– 359There is no corresponding record for this reference.
- 46Kirby, K. A.; Marchand, B.; Ong, Y. T.; Ndongwe, T. P.; Hachiya, A.; Michailidis, E.; Leslie, M. D.; Sietsema, D. V.; Fetterly, T. L.; Dorst, C. A.; Singh, K.; Wang, Z.; Parniak, M. A.; Sarafianos, S. G. Structural and inhibition studies of the RNase H function of xenotropic murine leukemia virus-related virus reverse transcriptase Antimicrob. Agents Chemother. 2012, 56, 2048– 2061There is no corresponding record for this reference.
- 47Michailidis, E.; Marchand, B.; Kodama, E. N.; Singh, K.; Matsuoka, M.; Kirby, K. A.; Ryan, E. M.; Sawani, A. M.; Nagy, E.; Ashida, N.; Mitsuya, H.; Parniak, M. A.; Sarafianos, S. G. Mechanism of inhibition of HIV-1 reverse transcriptase by 4′-ethynyl-2-fluoro-2′-deoxyadenosine triphosphate, a translocation-defective reverse transcriptase inhibitor J. Biol. Chem. 2009, 284, 35681– 35691There is no corresponding record for this reference.
- 48Wang, Z.; Bennett, E. M.; Wilson, D. J.; Salomon, C.; Vince, R. Rationally designed dual inhibitors of HIV reverse transcriptase and integrase J. Med. Chem. 2007, 50, 3416– 3419There is no corresponding record for this reference.
- 49Sirivolu, V. R.; Vernekar, S. K. V.; Ilina, T.; Myshakina, N. S.; Parniak, M. A.; Wang, Z. Q. Clicking 3′-Azidothymidine into Novel Potent Inhibitors of Human Immunodeficiency Virus J. Med. Chem. 2013, 56, 8765– 8780There is no corresponding record for this reference.
- 50HIV-1 reverse transcriptase with inhibitor; RCSB Protein Data Bank: Piscataway, NJ, 2010; http://www.rcsb.org/pdb/explore.do?structureId=3LP1, DOI: DOI: 10.2210/pdb3lp1/pdb .There is no corresponding record for this reference.
- 51Sastry, G. M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments J. Comput.-Aided Mol. Des. 2013, 27, 221– 23451Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichmentsSastry G Madhavi; Adzhigirey Matvey; Day Tyler; Annabhimoju Ramakrishna; Sherman WoodyJournal of computer-aided molecular design (2013), 27 (3), 221-34 ISSN:.Structure-based virtual screening plays an important role in drug discovery and complements other screening approaches. In general, protein crystal structures are prepared prior to docking in order to add hydrogen atoms, optimize hydrogen bonds, remove atomic clashes, and perform other operations that are not part of the x-ray crystal structure refinement process. In addition, ligands must be prepared to create 3-dimensional geometries, assign proper bond orders, and generate accessible tautomer and ionization states prior to virtual screening. While the prerequisite for proper system preparation is generally accepted in the field, an extensive study of the preparation steps and their effect on virtual screening enrichments has not been performed. In this work, we systematically explore each of the steps involved in preparing a system for virtual screening. We first explore a large number of parameters using the Glide validation set of 36 crystal structures and 1,000 decoys. We then apply a subset of protocols to the DUD database. We show that database enrichment is improved with proper preparation and that neglecting certain steps of the preparation process produces a systematic degradation in enrichments, which can be large for some targets. We provide examples illustrating the structural changes introduced by the preparation that impact database enrichment. While the work presented here was performed with the Protein Preparation Wizard and Glide, the insights and guidance are expected to be generalizable to structure-based virtual screening with other docking methods.
- 52(a) Schrödinger Release 2013-2: Schrödinger Suite 2013 Protein Preparation Wizard; Schrödinger, LLC: New York, 2013;There is no corresponding record for this reference.(b) Schrödinger Release 2013-2: Epik, version 2.5; Schrödinger, LLC: New York, 2013;There is no corresponding record for this reference.(c) Schrödinger Release 2013-2: Impact, version 6.0; Schrödinger, LLC: New York, 2013;There is no corresponding record for this reference.(d) Schrödinger Release 2013-2: Prime, version 3; Schrödinger, LLC: New York, 2013.There is no corresponding record for this reference.
- 53Jorgensen, W. L.; Maxwell, D. S.; TiradoRives, J. Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids J. Am. Chem. Soc. 1996, 118, 11225– 1123653Development and Testing of the OPLS All-Atom Force Field on Conformational Energetics and Properties of Organic LiquidsJorgensen, William L.; Maxwell, David S.; Tirado-Rives, JulianJournal of the American Chemical Society (1996), 118 (45), 11225-11236CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The parametrization and testing of the OPLS all-atom force field for org. mols. and peptides are described. Parameters for both torsional and nonbonded energetics have been derived, while the bond stretching and angle bending parameters have been adopted mostly from the AMBER all-atom force field. The torsional parameters were detd. by fitting to rotational energy profiles obtained from ab initio MO calcns. at the RHF/6-31G*//RHF/6-31G* level for more than 50 org. mols. and ions. The quality of the fits was high with av. errors for conformational energies of less than 0.2 kcal/mol. The force-field results for mol. structures are also demonstrated to closely match the ab initio predictions. The nonbonded parameters were developed in conjunction with Monte Carlo statistical mechanics simulations by computing thermodn. and structural properties for 34 pure org. liqs. including alkanes, alkenes, alcs., ethers, acetals, thiols, sulfides, disulfides, aldehydes, ketones, and amides. Av. errors in comparison with exptl. data are 2% for heats of vaporization and densities. The Monte Carlo simulations included sampling all internal and intermol. degrees of freedom. It is found that such non-polar and monofunctional systems do not show significant condensed-phase effects on internal energies in going from the gas phase to the pure liqs.
- 54Schrödinger Release 2013-2: Maestro, version 9.5; Schrödinger, LLC, New York, 2013.There is no corresponding record for this reference.
- 55Schrödinger Release 2013-2: LigPrep, version 2.7; Schrödinger, LLC, New York, 2013.There is no corresponding record for this reference.
Supporting Information
Supporting Information
Characterization data, including 1H NMR, 13C NMR, and HRMS data, of all intermediates. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.