51
|
Ahmad S, Waheed Y, Ismail S, Najmi MH, Ansari JK. Rational design of potent anti-COVID-19 main protease drugs: An extensive multi-spectrum in silico approach. J Mol Liq 2021; 330:115636. [PMID: 33612899 PMCID: PMC7879066 DOI: 10.1016/j.molliq.2021.115636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as a novel coronavirus and the etiological agent of global pandemic coronavirus disease (COVID-19) requires quick development of potential therapeutic strategies. Computer aided drug design approaches are highly efficient in identifying promising drug candidates among an available pool of biological active antivirals with safe pharmacokinetics. The main protease (MPro) enzyme of SARS-CoV-2 is considered key in virus production and its crystal structures are available at excellent resolution. This marks the enzyme as a good starting receptor to conduct an extensive structure-based virtual screening (SBVS) of ASINEX antiviral library for the purpose of uncovering valuable hits against SARS-CoV-2 MPro. A compound hit (BBB_26580140) was stand out in the screening process, as opposed to the control, as a potential inhibitor of SARS-CoV-2 MPro based on a combined approach of SBVS, drug likeness and lead likeness annotations, pharmacokinetics, molecular dynamics (MD) simulations, and end point MM-PBSA binding free energy methods. The lead was further used in ligand-based similarity search (LBSS) that found 33 similar compounds from the ChEMBL database. A set of three compounds (SCHEMBL12616233, SCHEMBL18616095, and SCHEMBL20148701), based on their binding affinity for MPro, was selected and analyzed using extensive MD simulation, hydrogen bond profiling, MM-PBSA, and WaterSwap binding free energy techniques. The compounds conformation with MPro show good stability after initial within active cavity moves, a rich intermolecular network of chemical interactions, and reliable relative and absolute binding free energies. Findings of the study suggested the use of BBB_26580140 lead and its similar analogs to be explored in vivo which might pave the path for rational drug discovery against SARS-CoV-2 MPro.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Foundation University Medical College, Foundation University Islamabad, Islambad, Pakistan
| | - Yasir Waheed
- Foundation University Medical College, Foundation University Islamabad, Islambad, Pakistan
| | - Saba Ismail
- Foundation University Medical College, Foundation University Islamabad, Islambad, Pakistan
| | - Muzammil Hasan Najmi
- Foundation University Medical College, Foundation University Islamabad, Islambad, Pakistan
| | - Jawad Khaliq Ansari
- Foundation University Medical College, Foundation University Islamabad, Islambad, Pakistan
| |
Collapse
|
52
|
Schleiff MA, Dhaware D, Sodhi JK. Recent advances in computational metabolite structure predictions and altered metabolic pathways assessment to inform drug development processes. Drug Metab Rev 2021; 53:173-187. [PMID: 33840322 DOI: 10.1080/03602532.2021.1910292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Many drug candidates fail during preclinical and clinical trials due to variable or unexpected metabolism which may lead to variability in drug efficacy or adverse drug reactions. The drug metabolism field aims to address this important issue from many angles which range from the study of drug-drug interactions, pharmacogenomics, computational metabolic modeling, and others. This manuscript aims to provide brief but comprehensive manuscript summaries highlighting the conclusions and scientific importance of seven exceptional manuscripts published in recent years within the field of drug metabolism. Two main topics within the field are reviewed: novel computational metabolic modeling approaches which provide complex outputs beyond site of metabolism predictions, and experimental approaches designed to discern the impacts of interindividual variability and species differences on drug metabolism. The computational approaches discussed provide novel outputs in metabolite structure and formation likelihood and/or extend beyond the saturated field of drug phase I metabolism, while the experimental metabolic pathways assessments aim to highlight the impacts of genetic polymorphisms and clinical animal model metabolic differences on human metabolism and subsequent health outcomes.
Collapse
Affiliation(s)
- Mary Alexandra Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Deepika Dhaware
- Biotransformation and ADME, Research and Development, Orion Corporation, Espoo, Finland
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
53
|
Karatas E, Foto E, Ertan-Bolelli T, Yalcin-Ozkat G, Yilmaz S, Ataei S, Zilifdar F, Yildiz I. Discovery of 5-(or 6)-benzoxazoles and oxazolo[4,5-b]pyridines as novel candidate antitumor agents targeting hTopo IIα. Bioorg Chem 2021; 112:104913. [PMID: 33945950 DOI: 10.1016/j.bioorg.2021.104913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/11/2021] [Accepted: 04/11/2021] [Indexed: 10/21/2022]
Abstract
Discovery of novel anticancer drugs which have low toxicity and high activity is very significant area in anticancer drug research and development. One of the important targets for cancer treatment research is topoisomerase enzymes. In order to make a contribution to this field, we have designed and synthesized some 5(or 6)-nitro-2-(substitutedphenyl)benzoxazole (1a-1r) and 2-(substitutedphenyl)oxazolo[4,5-b]pyridine (2a-2i) derivatives as novel candidate antitumor agents targeting human DNA topoisomerase enzymes (hTopo I and hTopo IIα). Biological activity results were found very promising for the future due to two compounds, 5-nitro-2-(4-butylphenyl)benzoxazole (1i) and 2-(4-butylphenyl)oxazolo[4,5-b]pyridine (2i), that inhibited hTopo IIα with 2 µM IC50 value. These two compounds were also found to be more active than reference drug etoposide. However, 1i and 2i did not show any satisfactory cyctotoxic activity on the HeLa, WiDR, A549, and MCF7 cancer cell lines. Moreover, molecular docking and molecular dynamic simulations studies for the most active compounds were applied in order to understand the mechanism of inhibition activity of hTopo IIα. In addition, in silico ADME/Tox studies were performed to predict drug-likeness and pharmacokinetic properties of all the tested compounds.
Collapse
Affiliation(s)
- Esin Karatas
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara, Turkey
| | - Egemen Foto
- Necmettin Erbakan University, Faculty of Science, Department of Biotechnology, Konya, Turkey
| | - Tugba Ertan-Bolelli
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara, Turkey
| | - Gozde Yalcin-Ozkat
- Ankara University, Biotechnology Institute, 0fef0 Yenimahalle, Ankara, Turkey; Recep Tayyip Erdogan University, Faculty of Engineering, Bioengineering Department, Rize, Turkey
| | - Serap Yilmaz
- Trakya University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Edirne, Turkey
| | - Sanaz Ataei
- Ankara University, Biotechnology Institute, 0fef0 Yenimahalle, Ankara, Turkey
| | - Fatma Zilifdar
- Selcuk University Faculty of Science, Department of Biochemistry, Konya, Turkey
| | - Ilkay Yildiz
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara, Turkey.
| |
Collapse
|
54
|
Santos J, Lobato L, Vale N. Clinical pharmacokinetic study of latrepirdine via in silico sublingual administration. In Silico Pharmacol 2021; 9:29. [PMID: 33898159 DOI: 10.1007/s40203-021-00083-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 03/09/2021] [Indexed: 10/21/2022] Open
Abstract
In recent decades, numerous in silico methodologies have been developed focused on the study of pharmacodynamic, pharmacokinetics and toxicological properties of drugs. The study of the pharmacokinetic behavior of new chemical entities is an essential part of the successful development of a new drug and Gastroplus™ is a simulation software used to predict the pharmacokinetic behavior of chemical entities. Latrepirdine is a drug that has been studied for Alzheimer's disease and Huntington's disease and later abandoned by the pharmaceutical industry already in the clinical trials because it has not demonstrated therapeutic efficacy. During this project, through Gastroplus™ simulations, it was possible to achieve predicted values of Cmax coincident with those found in clinical trials, showing its utility in the prediction of pharmacokinetic parameters. Besides, sublingual delivery has the potential to offer improved bioavailability by circumventing first-pass metabolism. This study used GastroPlus™ to simulate sublingual administration of latrepirdine and the results showed improvements in bioavailability and plasma concentrations achieved though this route of administration.
Collapse
Affiliation(s)
- Joana Santos
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine of University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.,Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Luísa Lobato
- Department of Nephrology, Centro Hospitalar Universitário do Porto, Hospital de Santo António, Porto, Portugal.,Unit for Multidisciplinary Research in Biomedicine, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine of University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal.,Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
55
|
Benzosuberene-sulfone analogues synthesis from Cedrus deodara oil and their therapeutic evaluation by computational analysis to treat type 2 diabetes. Bioorg Chem 2021; 112:104860. [PMID: 33839462 DOI: 10.1016/j.bioorg.2021.104860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/03/2021] [Accepted: 03/22/2021] [Indexed: 01/07/2023]
Abstract
Benzosuberene-sulfone (BSS) analogues have been semi-synthesized following green approaches from himachalenes, which has been extracted from essential oil of Cedrus deodara. In this process, benzosuberene in presence of different aryl or alkyl sodium sulfinates, I2 and potassium persulfate (K2S2O8) in acetonitrile-water solvent conditions gave BSS-analogues at room temperature. Under this reaction, a facile endocyclic β-H elimination has been noticed for BSS-analogues synthesis instead of vinyl sulfones and the reason may be due to its specific structure and electronic environment. The BSS-compounds were obtained with moderate to excellent yields under mild conditions. All the compounds were computationally subjected to drug likeliness and toxicity prediction studies. Further, the synthesized molecules were evaluated under in-silico studies for their binding affinity towards the native Peroxisome Proliferator-Activated Receptor Gamma (PPARG), and two PPARG mutants (R357A and V290M). Both the mutant forms of PPARG are deficient in eliciting a response to treatment with full and partial agonists. Our computational studies suggested that the molecule 3q performed better than the standard drug (Rosiglitazone) in all three protein structures. This implies that our suggested molecule could act as a more potent antagonist to native PPARG and could also be developed to treat type-2 diabetes patients with R357A and V290M mutations, which didn't elicit any response to currently available drugs in the market.
Collapse
|
56
|
Discovery of novel inhibitors against main protease (Mpro) of SARS-CoV-2 via virtual screening and biochemical evaluation. Bioorg Chem 2021; 110:104767. [PMID: 33667900 PMCID: PMC7903152 DOI: 10.1016/j.bioorg.2021.104767] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/24/2021] [Accepted: 02/17/2021] [Indexed: 01/19/2023]
Abstract
SARS-CoV-2 is the pathogen that caused the global COVID-19 outbreak in 2020. Promising progress has been made in developing vaccines and antiviral drugs. Antivirals medicines are necessary complements of vaccines for post-infection treatment. The main protease (Mpro) is an extremely important protease in the reproduction process of coronaviruses which cleaves pp1ab over more than 11 cleavage sites. In this work, two active main protease inhibitors were found via docking-based virtual screening and bioassay. The IC50 of compound VS10 was 0.20 μM, and the IC50 of compound VS12 was 1.89 μM. The finding in this work can be helpful to understand the interactions of main protease and inhibitors. The active candidates could be potential lead compounds for future drug design.
Collapse
|
57
|
Cardoso Santos C, Meuser Batista M, Inam Ullah A, Rama Krishna Reddy T, Soeiro MDNC. Drug screening using shape-based virtual screening and in vitro experimental models of cutaneous Leishmaniasis. Parasitology 2021; 148:98-104. [PMID: 33023678 PMCID: PMC11010133 DOI: 10.1017/s0031182020001900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 01/05/2023]
Abstract
Cutaneous leishmaniasis (CL) is one of the most disregarded tropical neglected disease with the occurrence of self-limiting ulcers and triggering mucosal damage and stigmatizing scars, leading to huge public health problems and social negative impacts. Pentavalent antimonials are the first-line drug for CL treatment for over 70 years and present several drawbacks in terms of safety and efficacy. Thus, there is an urgent need to search for non-invasive, non-toxic and potent drug candidates for CL. In this sense, we have implemented a shape-based virtual screening approach and identified a set of 32 hit compounds. In vitro phenotypic screenings were conducted using these hit compounds to check their potential leishmanicidal effect towards Leishmania amazonensis (L. amazonensis). Two (Cp1 and Cp2) out of the 32 compounds revealed promising antiparasitic activities, exhibiting considerable potency against intracellular amastigotes present in peritoneal macrophages (IC50 values of 9.35 and 7.25 μm, respectively). Also, a sterile cidality profile was reached at 20 μm after 48 h of incubation, besides a reasonable selectivity (≈8), quite similarly to pentamidine, a diamidine still in use clinically for leishmaniasis. Cp1 with an oxazolo[4,5-b]pyridine scaffold and Cp2 with benzimidazole scaffold could be developed by lead optimization studies to enhance their leishmanicidal potency.
Collapse
Affiliation(s)
- Camila Cardoso Santos
- Laboratory of Cellular Biology (LBC), Oswaldo Cruz Institute (IOC/FIOCRUZ), CEP21040-360, Rio de Janeiro, RJ, Brazil
| | - Marcos Meuser Batista
- Laboratory of Cellular Biology (LBC), Oswaldo Cruz Institute (IOC/FIOCRUZ), CEP21040-360, Rio de Janeiro, RJ, Brazil
| | - Asma Inam Ullah
- The Medicines Research Group, School of Health, Sport and Bioscience, College of Applied Health and Communities, University of East London, Stratford Campus, Water Lane, London, UK
| | - Tummala Rama Krishna Reddy
- The Medicines Research Group, School of Health, Sport and Bioscience, College of Applied Health and Communities, University of East London, Stratford Campus, Water Lane, London, UK
| | | |
Collapse
|
58
|
|
59
|
Bhardwaj VK, Singh R, Das P, Purohit R. Evaluation of acridinedione analogs as potential SARS-CoV-2 main protease inhibitors and their comparison with repurposed anti-viral drugs. Comput Biol Med 2020; 128:104117. [PMID: 33217661 PMCID: PMC7659809 DOI: 10.1016/j.compbiomed.2020.104117] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/01/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022]
Abstract
Background The main protease (Mpro) of SARS-CoV-2 is involved in the processing of vital polypeptides required for viral genome replication and transcription and is one of the best-characterized targets to inhibit the progression of SARS-CoV-2 in infected individuals. Methods We screened a set of novel classes of acridinediones molecules to efficiently bind and inhibit the activity of the SARS-CoV-2 by targeting the Mpro. The repurposed FDA-approved antivirals were taken as standard molecules for this study. Long term (1.1 μs) MD simulations were performed to analyze the conformational space of the binding pocket of Mpro bound to the selected molecules. Results The molecules DSPD-2 and DSPD-6 showed more favorable MM-PBSA interaction energies and were seated more deeply inside the binding pocket of Mpro than the topmost antiviral drug (Saquinavir). Moreover, DSPD-5 also exhibited comparable binding energy to Saquinavir. The analysis of per residue contribution energy and SASA studies indicated that the molecules showed efficient binding by targeting the S1 subsite of the Mpro binding pocket. Conclusion The DSPD-2, DSPD-6, and DSPD-5 could be developed as potential inhibitors of SARS-CoV-2. Moreover, we suggest that targeting molecules to bind effectively to the S1 subsite could potentially increase the binding of molecules to the SARS-CoV-2 Mpro. A robust computational strategy applied to identify the potential lead for COVID-19. Repurposed FDA approved antiviral drugs were compared with a set of acridinedione analogs against Mpro of SARS-CoV-2. The acridinedione analogs have acceptable ADMET values and low toxicity profile. In-house synthesized acridinedione analogs showed good amount of interaction with Mpro of SARS-CoV-2.
Collapse
Affiliation(s)
- Vijay Kumar Bhardwaj
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India; Biotechnology Division, CSIR-IHBT, Palampur, HP, 176061, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP, 176061, India
| | - Rahul Singh
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India; Biotechnology Division, CSIR-IHBT, Palampur, HP, 176061, India
| | - Pralay Das
- Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP, 176061, India; Natural Product Chemistry and Process Development, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP, 176061, India; Biotechnology Division, CSIR-IHBT, Palampur, HP, 176061, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP, 176061, India.
| |
Collapse
|
60
|
Singh R, Bhardwaj VK, Sharma J, Das P, Purohit R. Discovery and in silico evaluation of aminoarylbenzosuberene molecules as novel checkpoint kinase 1 inhibitor determinants. Genomics 2020; 113:707-715. [PMID: 33065246 DOI: 10.1016/j.ygeno.2020.10.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 10/01/2020] [Indexed: 01/12/2023]
Abstract
Checkpoint kinase 1 (CHK1) is an essential kinase with a critical function in cell cycle arrest. Several potent inhibitors targeting CHK1 have been published, but most of them have failed in clinical trials. Acknowledging the emerging consequence of CHK1 inhibitors in medication of cancer, there is a demand for widening the chemical range of CHK1 inhibitors. In this research, we considered a set of in-house plant based semi-synthetic aminoarylbenzosuberene molecules as potential CHK1 inhibitors. Based on a combined computational research that consolidates molecular docking and binding free energy computations we recognized the crucial determinants for their receptor binding. The drug likeness of these molecules were also scrutinized based on their toxicity and bioavailibilty profile. The computational strategy indicates that the Bch10 could be regarded as a potential CHK1 inhibitor in comparison with top five co-crystallize molecules. Bch10 signifies a promising outlet for the development of potent inhibitors for CHK1.
Collapse
Affiliation(s)
- Rahul Singh
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India
| | - Vijay Kumar Bhardwaj
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP 176061, India
| | - Jatin Sharma
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India
| | - Pralay Das
- Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP 176061, India; Natural Product Chemistry and Process Development, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, HP 176061, India; Biotechnology division, CSIR-IHBT, Palampur, HP 176061, India; Academy of Scientific & Innovative Research (AcSIR), CSIR-IHBT Campus, Palampur, HP 176061, India.
| |
Collapse
|
61
|
Wilt S, Kodani S, Le TNH, Nguyen L, Vo N, Ly T, Rodriguez M, Hudson PK, Morisseau C, Hammock BD, Pecic S. Development of multitarget inhibitors for the treatment of pain: Design, synthesis, biological evaluation and molecular modeling studies. Bioorg Chem 2020; 103:104165. [PMID: 32891856 DOI: 10.1016/j.bioorg.2020.104165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 11/30/2022]
Abstract
Multitarget-directed ligands are a promising class of drugs for discovering innovative new therapies for difficult to treat diseases. In this study, we designed dual inhibitors targeting the human fatty acid amide hydrolase (FAAH) enzyme and human soluble epoxide hydrolase (sEH) enzyme. Targeting both of these enzymes concurrently with single target inhibitors synergistically reduces inflammatory and neuropathic pain; thus, dual FAAH/sEH inhibitors are likely to be powerful analgesics. Here, we identified the piperidinyl-sulfonamide moiety as a common pharmacophore and optimized several inhibitors to have excellent inhibition profiles on both targeted enzymes simultaneously. In addition, several inhibitors show good predicted pharmacokinetic properties. These results suggest that this series of inhibitors has the potential to be further developed as new lead candidates and therapeutics in pain management.
Collapse
Affiliation(s)
- Stephanie Wilt
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Sean Kodani
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| | - Thanh N H Le
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Lato Nguyen
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Nghi Vo
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Tanya Ly
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Mark Rodriguez
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Paula K Hudson
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Christophe Morisseau
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| | - Bruce D Hammock
- Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States.
| |
Collapse
|
62
|
Biological evaluation and pharmacokinetic profiling of a coumarin-benzothiazole hybrid as a new scaffold for human gliomas. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
63
|
Olanlokun JO, Olotu FA, Idowu OT, Agoni C, David MO, Soliman M, Olorunsogo OO. In vitro, in silico studies of newly isolated tetrahydro-4-(7-hydroxy-10-methoxy-6, 14-dimethyl-15-m-tolylpentadec-13-enyl) pyran-2-one and isobutyryl acetate compounds from Alstonia boonei stem bark. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
64
|
Göller AH, Kuhnke L, Montanari F, Bonin A, Schneckener S, Ter Laak A, Wichard J, Lobell M, Hillisch A. Bayer's in silico ADMET platform: a journey of machine learning over the past two decades. Drug Discov Today 2020; 25:1702-1709. [PMID: 32652309 DOI: 10.1016/j.drudis.2020.07.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022]
Abstract
Over the past two decades, an in silico absorption, distribution, metabolism, and excretion (ADMET) platform has been created at Bayer Pharma with the goal to generate models for a variety of pharmacokinetic and physicochemical endpoints in early drug discovery. These tools are accessible to all scientists within the company and can be a useful in assisting with the selection and design of novel leads, as well as the process of lead optimization. Here. we discuss the development of machine-learning (ML) approaches with special emphasis on data, descriptors, and algorithms. We show that high company internal data quality and tailored descriptors, as well as a thorough understanding of the experimental endpoints, are essential to the utility of our models. We discuss the recent impact of deep neural networks and show selected application examples.
Collapse
Affiliation(s)
- Andreas H Göller
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 42096 Wuppertal, Germany
| | - Lara Kuhnke
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 13342 Berlin, Germany
| | - Floriane Montanari
- Bayer AG, Pharmaceuticals, R&D, Machine Learning Research, 13342 Berlin, Germany
| | - Anne Bonin
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 42096 Wuppertal, Germany
| | | | - Antonius Ter Laak
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 13342 Berlin, Germany
| | - Jörg Wichard
- Bayer AG, Pharmaceuticals, R&D, Genetic Toxicology, 13342 Berlin, Germany
| | - Mario Lobell
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 42096 Wuppertal, Germany
| | - Alexander Hillisch
- Bayer AG, Pharmaceuticals, R&D, Computational Molecular Design, 42096 Wuppertal, Germany.
| |
Collapse
|
65
|
Junaid A, Lim FPL, Tiekink ERT, Dolzhenko AV. Design, synthesis, and biological evaluation of new 6, N 2-diaryl-1,3,5-triazine-2,4-diamines as anticancer agents selectively targeting triple negative breast cancer cells. RSC Adv 2020; 10:25517-25528. [PMID: 35518627 PMCID: PMC9055250 DOI: 10.1039/d0ra04970k] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/28/2020] [Indexed: 12/24/2022] Open
Abstract
New 6,N2-diaryl-1,3,5-triazine-2,4-diamines were designed using the 3D-QSAR model developed earlier. These compounds were prepared and their antiproliferative activity was evaluated against three breast cancer cell lines (MDA-MB231, SKBR-3 and MCF-7) and non-cancerous MCF-10A epithelial breast cells. The synthesized compounds demonstrated selective antiproliferative activity against triple negative MDA-MB231 breast cancer cells. The most active compound in the series inhibited MDA-MB231 breast cancer cell growth with a GI50 value of 1 nM. None of the tested compounds significantly affected the growth of the normal breast cells. The time-dependent cytotoxic effect, observed when cytotoxicity was assessed at different time intervals after the treatment, and morphological features, observed in the fluorescence microscopy and live cell imaging experiments, suggested apoptosis as the main pathway for the antiproliferative activity of these compounds against MDA-MB231 cells. New highly potent and selective 6,N2-diaryl-1,3,5-triazine-2,4-diamines were designed and prepared using the 3D-QSAR model developed earlier.![]()
Collapse
Affiliation(s)
- Ahmad Junaid
- School of Pharmacy, Monash University Malaysia Jalan Lagoon Selatan Bandar Sunway Selangor Darul Ehsan 47500 Malaysia
| | - Felicia Phei Lin Lim
- School of Pharmacy, Monash University Malaysia Jalan Lagoon Selatan Bandar Sunway Selangor Darul Ehsan 47500 Malaysia
| | - Edward R T Tiekink
- Research Centre for Crystalline Materials, School of Science and Technology, Sunway University 5 Jalan Universiti Bandar Sunway Selangor Darul Ehsan 47500 Malaysia
| | - Anton V Dolzhenko
- School of Pharmacy, Monash University Malaysia Jalan Lagoon Selatan Bandar Sunway Selangor Darul Ehsan 47500 Malaysia .,School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University GPO Box U1987 Perth Western Australia 6845 Australia
| |
Collapse
|
66
|
Shen J, Nicolaou CA. Molecular property prediction: recent trends in the era of artificial intelligence. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 32-33:29-36. [PMID: 33386091 DOI: 10.1016/j.ddtec.2020.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/10/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022]
Abstract
Artificial intelligence (AI) has become a powerful tool in many fields, including drug discovery. Among various AI applications, molecular property prediction can have more significant immediate impact to the drug discovery process since most algorithms and methods use predicted properties to evaluate, select, and generate molecules. Herein, we provide a brief review of the state-of-art molecular property prediction methodologies and discuss examples reported recently. We highlight key techniques that have been applied to molecular property prediction such as learned representation, multi-task learning, transfer learning, and federated learning. We also point out some critical but less discussed issues such as data set quality, benchmark, model performance evaluation, and prediction confidence quantification.
Collapse
Affiliation(s)
- Jie Shen
- Advanced Analytics and Data Sciences, Eli Lilly and Company, Indianapolis, IN 46285, United States.
| | - Christos A Nicolaou
- Discovery Chemistry Research & Technologies, Eli Lilly and Company, Indianapolis, IN 46285, United States.
| |
Collapse
|
67
|
Davies M, Jones RDO, Grime K, Jansson-Löfmark R, Fretland AJ, Winiwarter S, Morgan P, McGinnity DF. Improving the Accuracy of Predicted Human Pharmacokinetics: Lessons Learned from the AstraZeneca Drug Pipeline Over Two Decades. Trends Pharmacol Sci 2020; 41:390-408. [PMID: 32359836 DOI: 10.1016/j.tips.2020.03.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 01/15/2023]
Abstract
During drug discovery and prior to the first human dose of a novel candidate drug, the pharmacokinetic (PK) behavior of the drug in humans is predicted from preclinical data. This helps to inform the likelihood of achieving therapeutic exposures in early clinical development. Once clinical data are available, the observed human PK are compared with predictions, providing an opportunity to assess and refine prediction methods. Application of best practice in experimental data generation and predictive methodologies, and a focus on robust mechanistic understanding of the candidate drug disposition properties before nomination to clinical development, have led to maximizing the probability of successful PK predictions so that 83% of AstraZeneca drug development projects progress in the clinic with no PK issues; and 71% of key PK parameter predictions [64% of area under the curve (AUC) predictions; 78% of maximum concentration (Cmax) predictions; and 70% of half-life predictions] are accurate to within twofold. Here, we discuss methods to predict human PK used by AstraZeneca, how these predictions are assessed and what can be learned from evaluating the predictions for 116 candidate drugs.
Collapse
Affiliation(s)
- Michael Davies
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
| | - Rhys D O Jones
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Ken Grime
- DMPK, Research and Early Development, Respiratory, Inflammation and Autoimmune, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson-Löfmark
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Adrian J Fretland
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Susanne Winiwarter
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Paul Morgan
- Mechanistic Safety and ADME Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Dermot F McGinnity
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
68
|
Griffen EJ, Dossetter AG, Leach AG. Chemists: AI Is Here; Unite To Get the Benefits. J Med Chem 2020; 63:8695-8704. [DOI: 10.1021/acs.jmedchem.0c00163] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Edward J. Griffen
- MedChemica Ltd., Alderley Park, Macclesfield, Cheshire SK10 4TG, U.K
| | | | - Andrew G. Leach
- MedChemica Ltd., Alderley Park, Macclesfield, Cheshire SK10 4TG, U.K
| |
Collapse
|
69
|
Russell LE, Schleiff MA, Gonzalez E, Bart AG, Broccatelli F, Hartman JH, Humphreys WG, Lauschke VM, Martin I, Nwabufo C, Prasad B, Scott EE, Segall M, Takahashi R, Taub ME, Sodhi JK. Advances in the study of drug metabolism - symposium report of the 12th Meeting of the International Society for the Study of Xenobiotics (ISSX). Drug Metab Rev 2020; 52:395-407. [PMID: 32456484 DOI: 10.1080/03602532.2020.1765793] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The 12th International Society for the Study of Xenobiotics (ISSX) meeting, held in Portland, OR, USA from July 28 to 31, 2019, was attended by diverse members of the pharmaceutical sciences community. The ISSX New Investigators Group provides learning and professional growth opportunities for student and early career members of ISSX. To share meeting content with those who were unable to attend, the ISSX New Investigators herein elected to highlight the "Advances in the Study of Drug Metabolism" symposium, as it engaged attendees with diverse backgrounds. This session covered a wide range of current topics in drug metabolism research including predicting sites and routes of metabolism, metabolite identification, ligand docking, and medicinal and natural products chemistry, and highlighted approaches complemented by computational modeling. In silico tools have been increasingly applied in both academic and industrial settings, alongside traditional and evolving in vitro techniques, to strengthen and streamline pharmaceutical research. Approaches such as quantum mechanics simulations facilitate understanding of reaction energetics toward prediction of routes and sites of drug metabolism. Furthermore, in tandem with crystallographic and orthogonal wet lab techniques for structural validation of drug metabolizing enzymes, in silico models can aid understanding of substrate recognition by particular enzymes, identify metabolic soft spots and predict toxic metabolites for improved molecular design. Of note, integration of chemical synthesis and biosynthesis using natural products remains an important approach for identifying new chemical scaffolds in drug discovery. These subjects, compiled by the symposium organizers, presenters, and the ISSX New Investigators Group, are discussed in this review.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Mary Alexandra Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric Gonzalez
- Division of Pre-Clinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, Bethesda, MD, USA.,Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Aaron G Bart
- Program in Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Fabio Broccatelli
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Jessica H Hartman
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Emily E Scott
- Program in Biophysics, University of Michigan, Ann Arbor, MI, USA.,Department of Medicinal Chemistry and Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT, USA
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
70
|
Norinder U, Spjuth O, Svensson F. Using Predicted Bioactivity Profiles to Improve Predictive Modeling. J Chem Inf Model 2020; 60:2830-2837. [PMID: 32374618 DOI: 10.1021/acs.jcim.0c00250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Predictive modeling is a cornerstone in early drug development. Using information for multiple domains or across prediction tasks has the potential to improve the performance of predictive modeling. However, aggregating data often leads to incomplete data matrices that might be limiting for modeling. In line with previous studies, we show that by generating predicted bioactivity profiles, and using these as additional features, prediction accuracy of biological endpoints can be improved. Using conformal prediction, a type of confidence predictor, we present a robust framework for the calculation of these profiles and the evaluation of their impact. We report on the outcomes from several approaches to generate the predicted profiles on 16 datasets in cytotoxicity and bioactivity and show that efficiency is improved the most when including the p-values from conformal prediction as bioactivity profiles.
Collapse
Affiliation(s)
- Ulf Norinder
- Department of Computer and Systems Sciences, Stockholm University, Box 7003, SE-164 07 Kista, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden.,MTM Research Centre, School of Science and Technology, Örebro University, SE-70182 Örebro, Sweden
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden.,Science for Life Laboratory, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Fredrik Svensson
- The Alzheimer's Research UK University College London Drug Discovery Institute, The Cruciform Building, Gower Street, WC1E 6BT London, U.K
| |
Collapse
|
71
|
Willems H, De Cesco S, Svensson F. Computational Chemistry on a Budget: Supporting Drug Discovery with Limited Resources. J Med Chem 2020; 63:10158-10169. [DOI: 10.1021/acs.jmedchem.9b02126] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Henriëtte Willems
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, U.K
| | - Stephane De Cesco
- Alzheimer’s Research UK Oxford Drug Discovery Institute, University of Oxford, NDM Research Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7FZ, U.K
| | - Fredrik Svensson
- Alzheimer’s Research UK UCL Drug Discovery Institute, University College London, The Cruciform Building, Gower Street, London WC1E 6BT, U.K
| |
Collapse
|
72
|
Fundamental aspects of DMPK optimization of targeted protein degraders. Drug Discov Today 2020; 25:969-982. [PMID: 32298797 DOI: 10.1016/j.drudis.2020.03.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/30/2022]
Abstract
Targeted protein degraders are an emerging modality. Their properties fall outside the traditional small-molecule property space and are in the 'beyond rule of 5' space. Consequently, drug discovery programs focused on developing orally bioavailable degraders are expected to face complex drug metabolism and pharmacokinetics (DMPK) challenges compared with traditional small molecules. Nevertheless, little information is available on the DMPK optimization of oral degraders. Therefore, in this review, we discuss our experience of these DMPK optimization challenges and present methodologies and strategies to overcome the hurdles dealing with this new small-molecule modality, specifically in developing oral degraders to treat cancer.
Collapse
|
73
|
Model-Informed Drug Discovery and Development Strategy for the Rapid Development of Anti-Tuberculosis Drug Combinations. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10072376] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The increasing emergence of drug-resistant tuberculosis requires new effective and safe drug regimens. However, drug discovery and development are challenging, lengthy and costly. The framework of model-informed drug discovery and development (MID3) is proposed to be applied throughout the preclinical to clinical phases to provide an informative prediction of drug exposure and efficacy in humans in order to select novel anti-tuberculosis drug combinations. The MID3 includes pharmacokinetic-pharmacodynamic and quantitative systems pharmacology models, machine learning and artificial intelligence, which integrates all the available knowledge related to disease and the compounds. A translational in vitro-in vivo link throughout modeling and simulation is crucial to optimize the selection of regimens with the highest probability of receiving approval from regulatory authorities. In vitro-in vivo correlation (IVIVC) and physiologically-based pharmacokinetic modeling provide powerful tools to predict pharmacokinetic drug-drug interactions based on preclinical information. Mechanistic or semi-mechanistic pharmacokinetic-pharmacodynamic models have been successfully applied to predict the clinical exposure-response profile for anti-tuberculosis drugs using preclinical data. Potential pharmacodynamic drug-drug interactions can be predicted from in vitro data through IVIVC and pharmacokinetic-pharmacodynamic modeling accounting for translational factors. It is essential for academic and industrial drug developers to collaborate across disciplines to realize the huge potential of MID3.
Collapse
|
74
|
Anasir MI, Ramanathan B, Poh CL. Structure-Based Design of Antivirals against Envelope Glycoprotein of Dengue Virus. Viruses 2020; 12:v12040367. [PMID: 32225021 PMCID: PMC7232406 DOI: 10.3390/v12040367] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Dengue virus (DENV) presents a significant threat to global public health with more than 500,000 hospitalizations and 25,000 deaths annually. Currently, there is no clinically approved antiviral drug to treat DENV infection. The envelope (E) glycoprotein of DENV is a promising target for drug discovery as the E protein is important for viral attachment and fusion. Understanding the structure and function of DENV E protein has led to the exploration of structure-based drug discovery of antiviral compounds and peptides against DENV infections. This review summarizes the structural information of the DENV E protein with regards to DENV attachment and fusion. The information enables the development of antiviral agents through structure-based approaches. In addition, this review compares the potency of antivirals targeting the E protein with the antivirals targeting DENV multifunctional enzymes, repurposed drugs and clinically approved antiviral drugs. None of the current DENV antiviral candidates possess potency similar to the approved antiviral drugs which indicates that more efforts and resources must be invested before an effective DENV drug materializes.
Collapse
Affiliation(s)
- Mohd Ishtiaq Anasir
- Center for Virus and Vaccine Research, School of Science and Technology, Sunway University, Kuala Lumpur, Selangor 47500, Malaysia;
| | - Babu Ramanathan
- Department of Biological Sciences, School of Science and Technology, Sunway University, Kuala Lumpur, Selangor 47500, Malaysia;
| | - Chit Laa Poh
- Center for Virus and Vaccine Research, School of Science and Technology, Sunway University, Kuala Lumpur, Selangor 47500, Malaysia;
- Correspondence: ; Tel.: +60-3-7491-8622; Fax: +60-3-5635-8633
| |
Collapse
|
75
|
Bonaccorso C, Naletova I, Satriano C, Spampinato G, Barresi V, Fortuna CG. New Di(heteroaryl)ethenes as Apoptotic Anti‐proliferative Agents Towards Breast Cancer: Design, One‐Pot Synthesis and In Vitro Evaluation. ChemistrySelect 2020. [DOI: 10.1002/slct.201903502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Carmela Bonaccorso
- Laboratorio di Modellistica Molecolare e dei Composti Eterociclici (ModHet) Dipartimento di Scienze Chimiche Università degli Studi di Catania Viale A. Doria 6 95125 Catania Italy
| | - Irina Naletova
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB) Via Celso Ulpiani, 27 70126 Bari, Italy
| | - Cristina Satriano
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB) Via Celso Ulpiani, 27 70126 Bari, Italy
- Laboratorio di NanobioInterfacce Ibride (NHIL) Dipartimento di Scienze Chimiche Università degli Studi di Catania Viale A. Doria 6 95125 Catania Italy
| | - Giorgia Spampinato
- Bio-nanotech Research and Innovation Tower (BRIT) Università degli Studi di Catania dsuakgbdshkj 95125 Catania Italy
- Dipartimento Scienze Biomediche e Biotecnologiche, Sez. Biochimica Medica Università degli Studi di Catania via S. Sofia 64 I-95125 Catania Italy
| | - Vincenza Barresi
- Bio-nanotech Research and Innovation Tower (BRIT) Università degli Studi di Catania dsuakgbdshkj 95125 Catania Italy
- Dipartimento Scienze Biomediche e Biotecnologiche, Sez. Biochimica Medica Università degli Studi di Catania via S. Sofia 64 I-95125 Catania Italy
| | - Cosimo G. Fortuna
- Laboratorio di Modellistica Molecolare e dei Composti Eterociclici (ModHet) Dipartimento di Scienze Chimiche Università degli Studi di Catania Viale A. Doria 6 95125 Catania Italy
| |
Collapse
|
76
|
Maharao N, Antontsev V, Wright M, Varshney J. Entering the era of computationally driven drug development. Drug Metab Rev 2020; 52:283-298. [PMID: 32083960 DOI: 10.1080/03602532.2020.1726944] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Historically, failure rates in drug development are high; increased sophistication and investment throughout the process has shifted the reasons for attrition, but the overall success rates have remained stubbornly and consistently low. Only 8% of new entities entering clinical testing gain regulatory approval, indicating that significant obstacles still exist for efficient therapeutic development. The continued high failure rate can be partially attributed to the inability to link drug exposure with the magnitude of observed safety and efficacy-related pharmacodynamic (PD) responses; frequently, this is a result of nonclinical models exhibiting poor prediction of human outcomes across a wide range of disease conditions, resulting in faulty evaluation of drug toxicology and efficacy. However, the increasing quality and standardization of experimental methods in preclinical stages of testing has created valuable data sets within companies that can be leveraged to further improve the efficiency and accuracy of preclinical prediction for both pharmacokinetics (PK) and PD. Models of Quantitative structure-activity relationships (QSAR), physiologically based pharmacokinetics (PBPK), and PK/PD relationships have also improved efficiency. Founded on a core understanding of biochemistry and physiological interactions of xenobiotics, these in silico methods have the potential to increase the probability of compound success in clinical trials. Integration of traditional computational methods with machine-learning approaches and existing internal pharma databases stands to make a fundamental impact on the speed and accuracy of predictions during the process of drug development and approval.
Collapse
|
77
|
Vo AH, Van Vleet TR, Gupta RR, Liguori MJ, Rao MS. An Overview of Machine Learning and Big Data for Drug Toxicity Evaluation. Chem Res Toxicol 2019; 33:20-37. [DOI: 10.1021/acs.chemrestox.9b00227] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Andy H. Vo
- Department of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Terry R. Van Vleet
- Department of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Rishi R. Gupta
- Information Research, Research and Development, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Michael J. Liguori
- Department of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Mohan S. Rao
- Department of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| |
Collapse
|
78
|
Bunally SB, Luscombe CN, Young RJ. Using Physicochemical Measurements to Influence Better Compound Design. SLAS DISCOVERY 2019; 24:791-801. [PMID: 31429385 DOI: 10.1177/2472555219859845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
During the past decade, the physicochemical quality of molecules under investigation at all stages of the drug discovery process has come under particular scrutiny. The issues associated with excessive lipophilicity and poor solubility in particular are many and varied, ranging from poor outcomes in screening campaigns to promiscuity, limited and/or poorly predictable pharmacokinetic exposure, and, ultimately, greater chances of clinical failure. In this review, contemporary methods to secure key measurements are described along with their relevance to understanding the behavior of molecules in environments pertinent to pharmacological activity. Together, the various measurements contribute to predictive models of both the physicochemical properties themselves and the outcomes they influence.
Collapse
Affiliation(s)
| | | | - Robert J Young
- 1 GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| |
Collapse
|
79
|
High-throughput hydrogen bond strength calculation and its applications in optimizing drug ADME properties. Future Med Chem 2019; 11:511-524. [PMID: 30892942 DOI: 10.4155/fmc-2018-0470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Modifying the molecule's intrinsic hydrogen bond strength (HBS) is a useful approach in optimizing its permeability and P-glycoprotein (P-gp) efflux. Quantum mechanics (QM) based computation has been utilized to estimate the molecular intrinsic HBS. Despite its usefulness, the computation is time consuming for a large set of molecules. METHODOLOGY/RESULTS We introduced a fragment-based high-throughput HBS calculation method and validated it with internal and external datasets. Examples have been presented where the P-gp efflux and permeability can be optimized by modulating calculated HBS. CONCLUSION The results will enable medicinal chemists to calculate HBS in a high-throughput manner while optimizing permeability and P-gp efflux. This will further improve the efficiency of balancing multiple properties during drug discovery process.
Collapse
|
80
|
Petito ES, Foster DJR, Ward MB, Sykes MJ. Molecular Modeling Approaches for the Prediction of Selected Pharmacokinetic Properties. Curr Top Med Chem 2019; 18:2230-2238. [PMID: 30569859 DOI: 10.2174/1568026619666181220105726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/22/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023]
Abstract
Poor profiles of potential drug candidates, including pharmacokinetic properties, have been acknowledged as a significant hindrance to the development of modern therapeutics. Contemporary drug discovery and development would be incomplete without the aid of molecular modeling (in-silico) techniques, allowing the prediction of pharmacokinetic properties such as clearance, unbound fraction, volume of distribution and bioavailability. As with all models, in-silico approaches are subject to their interpretability, a trait that must be balanced with accuracy when considering the development of new methods. The best models will always require reliable data to inform them, presenting significant challenges, particularly when appropriate in-vitro or in-vivo data may be difficult or time-consuming to obtain. This article seeks to review some of the key in-silico techniques used to predict key pharmacokinetic properties and give commentary on the current and future directions of the field.
Collapse
Affiliation(s)
- Emilio S Petito
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia Cancer Research Institute, Adelaide, South Australia 5001, Australia
| | - David J R Foster
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia Cancer Research Institute, Adelaide, South Australia 5001, Australia
| | - Michael B Ward
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia Cancer Research Institute, Adelaide, South Australia 5001, Australia
| | - Matthew J Sykes
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia Cancer Research Institute, Adelaide, South Australia 5001, Australia
| |
Collapse
|
81
|
Balaña-Fouce R, Pérez Pertejo MY, Domínguez-Asenjo B, Gutiérrez-Corbo C, Reguera RM. Walking a tightrope: drug discovery in visceral leishmaniasis. Drug Discov Today 2019; 24:1209-1216. [PMID: 30876846 DOI: 10.1016/j.drudis.2019.03.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/29/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
The current commitment of the pharma industry, nongovernmental organizations and academia to find better treatments against neglected tropical diseases should end decades of challenge caused by these global scourges. The initial result of these efforts has been the introduction of enhanced combinations of drugs, currently in clinical use, or formulations thereof. Phenotypic screening based on intracellular parasite infections has been revealed as the first key tool of antileishmanial drug discovery, because most first-in-class drugs entering Phase I trials were discovered this way. The professional commitment among stakeholders has enabled the availability of a plethora of new chemical entities that fit the target product profile for these diseases. However, the rate of hit discovery in leishmaniasis is far behind that for other neglected diseases. This review defends the need to develop new screening methods that consider the part played not only by intracellular parasites but also by the host's immune system to generate disease-relevant assays and improve clinical outcomes.
Collapse
Affiliation(s)
- Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - M Yolanda Pérez Pertejo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Bárbara Domínguez-Asenjo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Camino Gutiérrez-Corbo
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana, E-24071 León, Spain.
| |
Collapse
|
82
|
Docci L, Parrott N, Krähenbühl S, Fowler S. Application of New Cellular and Microphysiological Systems to Drug Metabolism Optimization and Their Positioning Respective to In Silico Tools. SLAS DISCOVERY 2019; 24:523-536. [PMID: 30817893 DOI: 10.1177/2472555219831407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New cellular model systems for drug metabolism applications, such as advanced 2D liver co-cultures, spheroids, and microphysiological systems (MPSs), offer exciting opportunities to reproduce human biology more closely in vitro with the aim of improving predictions of pharmacokinetics, drug-drug interactions, and efficacy. These advanced cellular systems have quickly become established for human intrinsic clearance determination and have been validated for several other absorption, distribution, metabolism, and excretion (ADME) applications. Adoption will be driven through the demonstration of clear added value, for instance, by more accurate and precise clearance predictions and by more reliable extrapolation of drug interaction potential leading to faster progression to pivotal proof-of-concept studies. New experimental systems are attractive when they can (1) increase experimental capacity, removing optimization bottlenecks; (2) improve measurement quality of ADME properties that impact pharmacokinetics; and (3) enable measurements to be made that were not previously possible, reducing risk in ADME prediction and candidate selection. As new systems become established, they will find their place in the repository of tools used at different stages of the research and development process, depending on the balance of value, throughput, and cost. In this article, we give a perspective on the integration of these new methodologies into ADME optimization during drug discovery, and the likely applications and impacts on drug development.
Collapse
Affiliation(s)
- Luca Docci
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland.,2 Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Neil Parrott
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | | | - Stephen Fowler
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
83
|
Broccatelli F, E.C.A Hop C, Wright M. Strategies to optimize drug half-life in lead candidate identification. Expert Opin Drug Discov 2019; 14:221-230. [DOI: 10.1080/17460441.2019.1569625] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Fabio Broccatelli
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Cornelis E.C.A Hop
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| | - Matthew Wright
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, USA
| |
Collapse
|
84
|
Zhou Y, Cahya S, Combs SA, Nicolaou CA, Wang J, Desai PV, Shen J. Exploring Tunable Hyperparameters for Deep Neural Networks with Industrial ADME Data Sets. J Chem Inf Model 2019; 59:1005-1016. [PMID: 30586300 DOI: 10.1021/acs.jcim.8b00671] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Deep learning has drawn significant attention in different areas including drug discovery. It has been proposed that it could outperform other machine learning algorithms, especially with big data sets. In the field of pharmaceutical industry, machine learning models are built to understand quantitative structure-activity relationships (QSARs) and predict molecular activities, including absorption, distribution, metabolism, and excretion (ADME) properties, using only molecular structures. Previous reports have demonstrated the advantages of using deep neural networks (DNNs) for QSAR modeling. One of the challenges while building DNN models is identifying the hyperparameters that lead to better generalization of the models. In this study, we investigated several tunable hyperparameters of deep neural network models on 24 industrial ADME data sets. We analyzed the sensitivity and influence of five different hyperparameters including the learning rate, weight decay for L2 regularization, dropout rate, activation function, and the use of batch normalization. This paper focuses on strategies and practices for DNN model building. Further, the optimized model for each data set was built and compared with the benchmark models used in production. Based on our benchmarking results, we propose several practices for building DNN QSAR models.
Collapse
Affiliation(s)
- Yadi Zhou
- Department of Chemistry and Biochemistry , Ohio University , Athens , Ohio 45701 , United States
| | | | | | | | | | | | | |
Collapse
|
85
|
Wenzel J, Matter H, Schmidt F. Predictive Multitask Deep Neural Network Models for ADME-Tox Properties: Learning from Large Data Sets. J Chem Inf Model 2019; 59:1253-1268. [DOI: 10.1021/acs.jcim.8b00785] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
86
|
Nasim MJ, Witek K, Kincses A, Abdin AY, Żesławska E, Marć MA, Gajdács M, Spengler G, Nitek W, Latacz G, Karczewska E, Kieć-Kononowicz K, Handzlik J, Jacob C. Pronounced activity of aromatic selenocyanates against multidrug resistant ESKAPE bacteria. NEW J CHEM 2019. [DOI: 10.1039/c9nj00563c] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Selenocyanates demonstrate pronounced activity against bacteria of the ESKAPE family, yeast and nematodes with limited cytotoxicity against human cells.
Collapse
|
87
|
Lombardo F, Berellini G, Obach RS. Trend Analysis of a Database of Intravenous Pharmacokinetic Parameters in Humans for 1352 Drug Compounds. Drug Metab Dispos 2018; 46:1466-1477. [PMID: 30115648 DOI: 10.1124/dmd.118.082966] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/09/2018] [Indexed: 11/22/2022] Open
Abstract
We report a trend analysis of human intravenous pharmacokinetic data on a data set of 1352 drugs. The aim in building this data set and its detailed analysis was to provide, as in the previous case published in 2008, an extended, robust, and accurate resource that could be applied by drug metabolism, clinical pharmacology, and medicinal chemistry scientists to a variety of scaling approaches. All in vivo data were obtained or derived from original references, either through the literature or regulatory agency reports, exclusively from studies utilizing intravenous administration. Plasma protein binding data were collected from other available sources to supplement these pharmacokinetic data. These parameters were analyzed concurrently with a range of physicochemical properties, and resultant trends and patterns within the data are presented. In addition, the date of first disclosure of each molecule was reported and the potential "temporal" impact on data trends was analyzed. The findings reported here are consistent with earlier described trends between pharmacokinetic behavior and physicochemical properties. Furthermore, the availability of a large data set of pharmacokinetic data in humans will be important to further pursue analyses of physicochemical properties, trends, and modeling efforts and should propel our deeper understanding (especially in terms of clearance) of the absorption, distribution, metabolism, and excretion behavior of drug compounds.
Collapse
Affiliation(s)
- Franco Lombardo
- Drug Metabolism and Bioanalysis Group, Alkermes Inc., Waltham, Massachusetts (F.L.); Computational Chemistry Group, Biogen Inc., Cambridge, Massachusetts (G.B.); and Pharmacokinetics Dynamics and Metabolism Department, Groton Laboratories, Pfizer Global Research and Development, Groton, Connecticut (R.S.O.)
| | - Giuliano Berellini
- Drug Metabolism and Bioanalysis Group, Alkermes Inc., Waltham, Massachusetts (F.L.); Computational Chemistry Group, Biogen Inc., Cambridge, Massachusetts (G.B.); and Pharmacokinetics Dynamics and Metabolism Department, Groton Laboratories, Pfizer Global Research and Development, Groton, Connecticut (R.S.O.)
| | - R Scott Obach
- Drug Metabolism and Bioanalysis Group, Alkermes Inc., Waltham, Massachusetts (F.L.); Computational Chemistry Group, Biogen Inc., Cambridge, Massachusetts (G.B.); and Pharmacokinetics Dynamics and Metabolism Department, Groton Laboratories, Pfizer Global Research and Development, Groton, Connecticut (R.S.O.)
| |
Collapse
|
88
|
Can we accelerate medicinal chemistry by augmenting the chemist with Big Data and artificial intelligence? Drug Discov Today 2018; 23:1373-1384. [DOI: 10.1016/j.drudis.2018.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/27/2018] [Accepted: 03/20/2018] [Indexed: 12/18/2022]
|
89
|
Hop P, Allgood B, Yu J. Geometric Deep Learning Autonomously Learns Chemical Features That Outperform Those Engineered by Domain Experts. Mol Pharm 2018; 15:4371-4377. [DOI: 10.1021/acs.molpharmaceut.7b01144] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Patrick Hop
- Numerate Inc., San Francisco, California 94107, United States
| | - Brandon Allgood
- Numerate Inc., San Francisco, California 94107, United States
| | - Jessen Yu
- Numerate Inc., San Francisco, California 94107, United States
| |
Collapse
|