Improving the Use of Ranking in Virtual Screening against HIV-1 Integrase with Triangular Numbers and Including Ligand Profiling with Antitargets

A delicate balance exists between a drug molecule's toxicity and its activity. Indeed, efficacy, toxicity, and side effect problems are a common cause for the termination of drug candidate compounds and development projects. To address this, an antitarget interaction profile is built and combined with virtual screening and cross docking for new inhibitors of HIV-1 integrase, in order to consider possible off-target interactions as early as possible in a drug or hit discovery program. New ranking techniques using triangular numbers improve ranking information on the compounds and recovery of known inhibitors into the top compounds using different docking programs. This improved ranking arises from using consensus of ranks between docking programs and ligand efficiencies to derive a new rank, instead of using absolute score values, or average of ranks. The triangular number rerank also allowed the objective combination of results from several protein targets or screen conditions and several programs. Triangular number reranking conserves more information than other reranking methods such as average of scores or averages of ranks. In addition, the use of triangular numbers for reranking makes possible the use of thresholds with a justified leeway based on the number of available known inhibitors, so that the majority of the compounds above the threshold in ranks compare to the compounds that have known experimentally determined biological activity. The battery of anti- or off-targets can be tailored to specific molecular or drug design challenges. In silico filters can thus be deployed in successive stages, for prefiltering, activity profiling, and for further analysis and triaging of libraries of compounds.

[1]  G. Cohen,et al.  Structure of the HIV-1 integrase catalytic domain complexed with an inhibitor: a platform for antiviral drug design. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[2]  A. Hopkins Network pharmacology: the next paradigm in drug discovery. , 2008, Nature chemical biology.

[3]  Christine Humblet,et al.  Computation of 3D queries for ROCS based virtual screens , 2009, J. Comput. Aided Mol. Des..

[4]  A. Hopkins,et al.  Ligand efficiency: a useful metric for lead selection. , 2004, Drug discovery today.

[5]  H. Doerr,et al.  CYTARABINE TREATMENT OF HUMAN T-LYMPHOID CELLS INDUCES DECREASED HIV-1 RECEPTOR EXPRESSION AND REDUCED HIV-1 SUSCEPTIBILITY , 2001, Nucleosides, nucleotides & nucleic acids.

[6]  Uko Maran,et al.  DrugLogit: Logistic Discrimination between Drugs and Nondrugs Including Disease-Specificity by Assigning Probabilities Based on Molecular Properties , 2012, J. Chem. Inf. Model..

[7]  Yuan Zhao,et al.  Computation of Octanol-Water Partition Coefficients by Guiding an Additive Model with Knowledge , 2007, J. Chem. Inf. Model..

[8]  Alfonso T. García-Sosa,et al.  Hydration Properties of Ligands and Drugs in Protein Binding Sites: Tightly-Bound, Bridging Water Molecules and Their Effects and Consequences on Molecular Design Strategies , 2013, J. Chem. Inf. Model..

[9]  David S. Wishart,et al.  DrugBank: a comprehensive resource for in silico drug discovery and exploration , 2005, Nucleic Acids Res..

[10]  Dima Kozakov,et al.  The structural basis of pregnane X receptor binding promiscuity. , 2009, Biochemistry.

[11]  P. Leeson,et al.  The influence of drug-like concepts on decision-making in medicinal chemistry , 2007, Nature Reviews Drug Discovery.

[12]  Roy J. Vaz,et al.  Antitargets : prediction and prevention of drug side effects , 2008 .

[13]  Anthony Nicholls,et al.  What do we know and when do we know it? , 2008, J. Comput. Aided Mol. Des..

[14]  Uko Maran,et al.  Disease‐Specific Differentiation Between Drugs and Non‐Drugs Using Principal Component Analysis of Their Molecular Descriptor Space , 2012, Molecular informatics.

[15]  Stephen R. Johnson,et al.  Molecular properties that influence the oral bioavailability of drug candidates. , 2002, Journal of medicinal chemistry.

[16]  Sergio Martí,et al.  A quantum mechanics/molecular mechanics study of the protein-ligand interaction for inhibitors of HIV-1 integrase. , 2007, Chemistry.

[17]  A. Zelenetz,et al.  Intensive chemotherapy with cyclophosphamide, doxorubicin, high‐dose methotrexate/ifosfamide, etoposide, and high‐dose cytarabine (CODOX‐M/IVAC) for human immunodeficiency virus–associated Burkitt lymphoma , 2003, Cancer.

[18]  I. Kuntz,et al.  The maximal affinity of ligands. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[19]  David S. Goodsell,et al.  Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function , 1998 .

[20]  Dale J. Kempf,et al.  A Human Immunodeficiency Virus Protease Inhibitor Is a Novel Functional Inhibitor of Human Pregnane X Receptor , 2008, Drug Metabolism and Disposition.

[21]  Simona Distinto,et al.  How To Optimize Shape-Based Virtual Screening: Choosing the Right Query and Including Chemical Information , 2009, J. Chem. Inf. Model..

[22]  John P. Overington,et al.  Can we rationally design promiscuous drugs? , 2006, Current opinion in structural biology.

[23]  Yvonne C. Martin,et al.  Let’s not forget tautomers , 2009, J. Comput. Aided Mol. Des..

[24]  B. Gazzard,et al.  The management of meningeal lymphoma in patients with HIV in the era of HAART: intrathecal depot cytarabine is effective and safe. , 2006, Blood.

[25]  Brian K. Shoichet,et al.  ZINC - A Free Database of Commercially Available Compounds for Virtual Screening , 2005, J. Chem. Inf. Model..

[26]  Uko Maran,et al.  Combined Approach Using Ligand Efficiency, Cross-Docking, and Antitarget Hits for Wild-Type and Drug-Resistant Y181C HIV-1 Reverse Transcriptase , 2011, J. Chem. Inf. Model..

[27]  M. Parker,et al.  Fragment‐Based Design of Ligands Targeting a Novel Site on the Integrase Enzyme of Human Immunodeficiency Virus 1 , 2011, ChemMedChem.

[28]  J. T. Metz,et al.  Ligand efficiency indices as guideposts for drug discovery. , 2005, Drug discovery today.

[29]  F. Bailly,et al.  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. , 2008, Journal of medicinal chemistry.

[30]  B. Shoichet,et al.  Molecular docking and ligand specificity in fragment-based inhibitor discovery. , 2009, Nature chemical biology.

[31]  Christopher L. McClendon,et al.  Reaching for high-hanging fruit in drug discovery at protein–protein interfaces , 2007, Nature.

[32]  John P. Overington,et al.  Probing the links between in vitro potency, ADMET and physicochemical parameters , 2011, Nature Reviews Drug Discovery.

[33]  William L. Jorgensen,et al.  Discovery of Wild-Type and Y181C Mutant Non-nucleoside HIV-1 Reverse Transcriptase Inhibitors Using Virtual Screening with Multiple Protein Structures , 2009, J. Chem. Inf. Model..

[34]  M. Congreve,et al.  A 'rule of three' for fragment-based lead discovery? , 2003, Drug discovery today.

[35]  Uko Maran,et al.  Drug efficiency indices for improvement of molecular docking scoring functions , 2010, J. Comput. Chem..

[36]  P. R. Montellano Hydrocarbon hydroxylation by cytochrome P450 enzymes. , 2010 .

[37]  E. García-Martín,et al.  Interethnic and Intraethnic Variability of CYP2C8 and CYP2C9 Polymorphisms in Healthy Individuals , 2012, Molecular Diagnosis & Therapy.

[38]  A. D. Rodrigues,et al.  Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes. , 1996, The Journal of pharmacology and experimental therapeutics.

[39]  D. Hazuda,et al.  Discovery of raltegravir, a potent, selective orally bioavailable HIV-integrase inhibitor for the treatment of HIV-AIDS infection. , 2008, Journal of medicinal chemistry.

[40]  Arthur J. Olson,et al.  AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading , 2009, J. Comput. Chem..

[41]  Carlos A. Montanari,et al.  Novel Application of 2D and 3D-Similarity Searches To Identify Substrates among Cytochrome P450 2C9, 2D6, and 3A4 , 2010, J. Chem. Inf. Model..

[42]  S. Montoto,et al.  Excellent immunological recovery following CODOX-M/IVAC, an effective intensive chemotherapy for HIV-associated Burkitt's lymphoma , 2010, AIDS.

[43]  Uko Maran,et al.  Docking and Virtual Screening Using Distributed Grid Technology , 2009 .

[44]  M. Congreve,et al.  Fragment-based lead discovery , 2004, Nature Reviews Drug Discovery.

[45]  P. Bork,et al.  Drug Target Identification Using Side-Effect Similarity , 2008, Science.

[46]  Peter G Schultz,et al.  A potent and highly selective sulfotransferase inhibitor. , 2002, Journal of the American Chemical Society.

[47]  Michael J. Keiser,et al.  Predicting new molecular targets for known drugs , 2009, Nature.

[48]  Tudor I. Oprea,et al.  Is There a Difference between Leads and Drugs? A Historical Perspective , 2001, J. Chem. Inf. Comput. Sci..

[49]  C. Salomon,et al.  Rationally designed dual inhibitors of HIV reverse transcriptase and integrase. , 2007, Journal of medicinal chemistry.