Next Article in Journal
Playing with Structural Parameters: Synthesis and Characterization of Two New Maltol-Based Ligands with Binding and Antineoplastic Properties
Next Article in Special Issue
Discovery of Novel Inhibitor for WNT/β-Catenin Pathway by Tankyrase 1/2 Structure-Based Virtual Screening
Previous Article in Journal
Synthetic Compounds with 2-Amino-1,3,4-Thiadiazole Moiety Against Viral Infections
Previous Article in Special Issue
Design of Disruptors of the Hsp90–Cdc37 Interface
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Large-Scale Virtual Screening Against the MET Kinase Domain Identifies a New Putative Inhibitor Type

1
Université de Lorraine, CNRS, Inria, LORIA, F-54000 Nancy, France
2
Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), UMR7288, Parc Scientifique de Luminy, 13009 Marseille, France
3
University of Lille, CNRS, UMR 8523, PhLAM-Physique des Lasers Atomes et Molécules, F-59000 Lille, France
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2020, 25(4), 938; https://doi.org/10.3390/molecules25040938
Submission received: 27 January 2020 / Revised: 13 February 2020 / Accepted: 14 February 2020 / Published: 19 February 2020
(This article belongs to the Special Issue Drug Discovery and Molecular Docking)

Abstract

:
By using an ensemble-docking strategy, we undertook a large-scale virtual screening campaign in order to identify new putative hits against the MET kinase target. Following a large molecular dynamics sampling of its conformational space, a set of 45 conformers of the kinase was retained as docking targets to take into account the flexibility of the binding site moieties. Our screening funnel started from about 80,000 chemical compounds to be tested in silico for their potential affinities towards the kinase binding site. The top 100 molecules selected—thanks to the molecular docking results—were further analyzed for their interactions, and 25 of the most promising ligands were tested for their ability to inhibit MET activity in cells. F0514-4011 compound was the most efficient and impaired this scattering response to HGF (Hepatocyte Growth Factor) with an IC 50 of 7.2 μ M. Interestingly, careful docking analysis of this molecule with MET suggests a possible conformation halfway between classical type-I and type-II MET inhibitors, with an additional region of interaction. This compound could therefore be an innovative seed to be repositioned from its initial antiviral purpose towards the field of MET inhibitors. Altogether, these results validate our ensemble docking strategy as a cost-effective functional method for drug development.

1. Introduction

MET receptor is a multifunctional receptor tyrosine kinase that plays a pivotal role in human development and tumorigenesis. Upon binding of its ligand HGF (Hepatocyte Growth Factor), MET triggers several intracellular signaling cascades, including MAPK and PI3K pathways, leading to various cellular responses, such as survival, proliferation, and migration, among others. MET activation can be driven in cancer by several mechanisms: HGF or MET overexpression, and also mutations [1].
Aberrant activation of MET signaling does not only affect cancer development and progression, but it also contributes to resistance against other cancer drugs [2,3,4,5,6,7,8,9,10,11]. Consequently, MET represents a pharmaceutically relevant target in anticancer drug design and has been the focus of several anticancer strategies [12,13,14,15,16,17,18]. Pioneer MET inhibitors such as SU11274, PHA665752, or AM7 were helpful for demonstrating the efficacy of MET inhibition to impair tumor growth in preclinical models. Then, further developments in the field led to the approval by the FDA of crizotinib and cabozantinib in the 2010s for treating non-small cell lung cancers and medullary thyroid cancers, respectively.
Even though promising results have been reported, the therapeutic activity of these drugs is still relative and efforts are required to identify new MET inhibitors with physicochemical properties optimized for clinical efficiency [19,20]. Moreover, new alterations in MET sequence have been recently identified, such as MET exon 14 skipping in lung cancers and the emergence of MET mutations in the kinase domain following treatment with MET inhibitors [21]. Novel inhibitor structures may possibly target these mutations with increased efficiency.
Designing new putative hits against MET therefore remains a valuable challenge to be tackled. In the present work, we carried out a virtual screening campaign in order to identify innovative compounds able to become new hits for further lead development. As MET plasticity upon ligand binding had been previously highlighted [22,23], we took into account this aspect for the molecular docking simulations. Indeed, MET can accommodate several distinct ligand binding modes and associated receptor conformations, a feature that is particular to the kinase family [24]. We reasoned that it should be taken into account for designing drugs with improved efficiency and selectivity profiles [25,26]. To be efficient, the molecular docking engines embedded within the virtual screening approaches must be adapted to handle such flexibility [27,28]. In the present work, we used the ensemble-docking strategy—previously recognized for its efficiency in drug design [29]—and show the benefit of an investigation using a large ensemble-docking on MET.
In previous medicinal chemistry works, we already identified novel MET inhibitors [30,31] and characterized the different MET ligand binding modes as shown by the stream of released X-ray data [32]. Here, we provide the results of a large-scale ensemble-docking investigation on MET, in which MET conformations are extended from available X-ray data to molecular dynamics and normal mode analysis. A limited number of candidate compounds were selected from the ensemble-docking results and one of them was subsequently validated experimentally as a possible new MET inhibitor, providing a valuable seed for further investigations.

2. Materials and Methods

2.1. Screened Chemical Libraries

The choice of an appropriated set of compounds to explore the virtual screening space is critical for assuming a good rate of success [33]. Today, millions of compounds can be selected for high-throughput screening, and a suitable selection strategy must be designed. In our case and according to previous success stories [34,35,36], we chose to use a set of libraries selected in order:
  • To use the highest possible chemical diversity, in order to cover a large spectrum of chemical structures [37,38,39,40];
  • To include kinase-targeted compounds, as such a choice is already proven to be successful [41,42];
  • To explore natural products, which are a promising source of anticancer drugs [43,44,45];
  • To take into account the repositioning of approved compounds, as drug repurposing presents an increasing interest in cancer research, by removing many costs associated with several steps of drug development [46,47,48,49]. Several proofs of concept are now available and a typical case of viral-to-cancer drug repositioning is gemcitabine with US patent No 4,808,614, aimed at treating viral infections, and the later-issued US No 5,464,826, which claims of its use to treat cancer. Therefore, we also considered chemical libraries dedicated to antiviral compounds.
According to the criteria listed above concerning the choice of the chemical libraries, we firstly downloaded around 200,000 compounds from the chemical providers listed in Table 1. After eliminating redundancies in compounds and in scaffolds to assume a large chemical diversity and in respect of general druglike definitions [50], we finally retained about 80,000 compounds for our screening campaign.

2.2. Selected MET Conformational Ensemble

In ensemble-based docking calculations, a well-suited choice of the protein target conformational sample is required to reproduce the protein plasticity and the possible induced-fit phenomena [51,52].
Concerning MET conformational flexibility, our previous exploration by molecular dynamics and normal mode simulations [22] was limited to the 26 PDB structures available at that time (Table 2). We have now extended this analysis by considering all the X-ray structures available for the wild-type MET in the PDB [53] deposited after 2012. From the additional structures found, only 19 were considered in this work (Table 3) as we discarded those where three regions were missing in the X-ray structure and those where the number of missing residues in a single region was larger than 20. This selection aimed to improve our protein ensemble sample by covering most of the kinase structural characteristics, such as the position of the c-helix (in or out) or of the aspartate-phenylalanine-glycine (DFG) motif (in or out) as defined in Kinametrix [54], thus covering most of the inhibitor type binding modes. 3D structures considered in this ensemble of 45 conformers looked representative of the diversity of MET structures, as shown from the dendrogram, the heat maps, and correspondence maps in Figure 1. These results obtained thanks to the Dali server [55] clearly illustrate how MET 3D structures used here are organized into several families covering most of the protein conformational space presently known.
To be in accordance with the 26 conformers coming from our previous work [22], the 19 added crystal structures were prepared and cleaned following the same protocol: missing residues, side chains, and hydrogens were added when necessary; unnecessary water molecules, ions, and additives were removed; basic and acidic side chains were ionized according to a pH set to 7. To consider possible binding sites fluctuations, short molecular dynamics (MD) simulations were undertaken for each of these 19 structures. For that purpose, these structures were placed in a solvent box of 80 Å and counter ions were added for electrostatic neutrality. NAMD [57] molecular dynamics program was used with the same CHARMM36 force field and same protocol as previously described [22]. After minimization and equilibrium steps (64,000 conjugate gradients and 1 ns MD, respectively), 10 ns of MD were recorded, with a frame length of 1 ps. These 19 MD trajectories were analyzed, and the most stable representative conformer was retained for each of them and added in the ensemble-docking set.

2.3. Description of the Ensemble-Docking Protocol

The ensemble-docking facility proposed in the GOLD docking program was used [58]. This GOLD feature evaluates different receptor conformations concurrently during the docking exploration. The protein ensemble used in this work thus contained 45 MET conformers (26 from our previous work and 19 added in this one). As these conformers must be superimposed before being used in GOLD ensemble-docking program, they were structurally aligned according to their conserved and most rigid secondary structure patterns, as previously described [22] and summarized in Table 4.
When docking an ensemble of conformations for a given protein, their binding sites must be defined using a method that is not conformer specific. In the present ensemble-GOLD version, as it was not possible to define the active site by a list of atoms or residues, the only way was to use the centroid of the binding cavity and a sphere radius around this point. Therefore, for each of the 45 aligned protein conformers used here, protein cavities and their center of mass were detected by the LIGSITE program [59]. From these data, we obtained an average center point as the ensemble binding site definition for GOLD. Figure 2 presents the position of this average center point within the 45 protein conformers. A radius of 20 Å was associated to this average point to define the binding cavity of each conformer in order to correctly encompass the receptor for all the conformations in the ensemble, including conformational variations around the center. We also verified that the resulting sphere was encompassing all groups of residues previously identified as potential interaction areas for MET ligands [32].
For each docking run, we used 50 starting poses/molecule for the GOLD generic algorithm. Tested compounds were ranked by the standard goldscore scoring function.

2.4. Computer Grid Facilities

Due to the massive calculations needed ( 80,000 molecules × 48 protein ensemble conformers × 50 poses/molecule), and considering the computing time to achieve only one run, we used the Grid5000 facility [60] providing the required computer power in order to distribute the calculations using the PVM framework embedded in GOLD. A total of 1300 cpus (mostly Xeons) with 4 GB RAM/core and infiniband connections were used for each run. The docking performances run around 300-docked ligands/ensemble/hour. The calculations were spread on the clusters using the same strategy as previously described [61].

2.5. Scattering Assays

The experimental protocols for measuring the potency of MET inhibitors are detailed in previous publications [30,62]. MDCK cells were preincubated with compounds overnight at 0.1–100 μ M concentrations at 37 °C in a humidified atmosphere of 5% CO 2 , followed by a 24 h stimulation with 20 ng/mL HGF (R&D Systems). Cells were further incubated at 37 °C in an atmosphere of 5% CO 2 for 24–48 h, washed with phosphate buffered saline (PBS; Gibco BRL), and fixed with 4% PFA (paraformaldehyde, Sigma). The quantification of scattering response was performed by counting the number of cells with scattered morphology in 30 independent colonies. The IC 50 corresponds to the concentration of compounds leading to a 50% inhibition of MET-triggered cell scattering.

3. Results

3.1. Preliminary Validation Concerning the GOLD Ensemble-Docking Protocol Used

The coordinates of the 45 aligned conformers and of the sphere representing their common binding sites constituted our ensemble-docking protein reference.
The first question here concerned the accuracy of this binding site definition compared to ones that are more classical. For that, we compared the docking results for some of the selected 45 MET conformers using three binding site definitions; namely, a residue list, an existing ligand, and the center point of the binding cavity, respectively. For each individual docking target, the three definitions provided almost the same rank and docking score for the associated PDB ligand (Table 5). Moreover, the poses of this ligand found using the three binding site definitions were similar to the pose found in the crystal structures, as illustrated with the example of the AM7 ligand on Figure 3.
The second question was related to the ability of the ensemble-docking process to retrieve a given PDB ligand to its PDB structure among the 45 ones. To evaluate that point, an ensemble-docking calculation was carried out on the 45 protein target conformers using a short chemical library built from their own 45 associated ligands (the list is given in Table 2 and Table 3). We checked whether we could associate the right PDB target for a given PDB ligand (with possibly similar rank, score and pose compared to the ones found for the individual target dockings) in the protein ensemble. This was achieved for almost 80% of the compounds (Table S1). For example, the KSA ligand was able to preferentially retrieve its original 1R0P partner among the ensemble of the 45 PDB conformers of the protein target.
From these results, it appeared that the ensemble-docking procedure we used was a satisfactory method to tackle multiple conformers docking and to achieve a valuable virtual screening.

3.2. Selection of Candidate Hits from the Virtual Screening Campaign

Once the screening campaign was achieved for the 80,000 compounds filtered from the chosen libraries, we kept the top-100 ranked compounds according to their GOLD scores (ranging from 100 to 114) for further analysis.
We started the docking analysis with the Life Chemicals compound F0725-0356 giving the best docking score of 114. A comparison between the X-ray complex 3EFK/MT4 structure and the MD_3EFK/F025-0356 one presented quite similar poses and protein/ligand interactions. Indeed, the most important residues known in MET interactions (namely, Met 1160 , Asp 1222 , Tyr 1159 ) were found in both complexes.
We next analyzed the protein-ligand interactions for the other top-100 compounds in order to compare them to the ones found in the 45 original PDB structures (Table 2 and Table 3). For that, we used the PLIP program [63] by focusing on two important interaction types: hydrogen bonds and π -stacking. Protein residues Met 1160 (45/45), Asp 1222 (34/45), and Lys 1110 (6/45) concentrated the vast majority of hydrogen bonds with ligands; while Tyr 1230 (25/45) and Phe 1223 (7/45) dealt with most of the π -stacking. In order to limit our biological tests on possible promising compounds, we eliminated from the top-100 list the molecules not presenting at least one hydrogen bond and one π -stacking from the ones described above in the PDB complexes.
After this filter, we retained only 41 compounds as satisfying these criteria. As most of these compounds came from the Life Chemical antiviral library and given the simplicity of comparing molecules from the same supplier, we decided to only test compounds from Life Chemicals. As some of these molecules were not available in stock from this provider, only the 25 compounds listed in Table 6 were finally kept to proceed further.

3.3. F0539-1482 and F0514-4011 Inhibit MET-Induced Cell Scattering

These 25 compounds were then experimentally tested for their ability to restrain MET-triggered biological activities. We previously efficiently screened compounds for their inhibitory properties towards MET-triggered biological responses by using cell scattering assays [31,64]. In particular, MDCK epithelial cells acquire a “scattered phenotype” after stimulation with MET ligand HGF.
Among the 25 tested compounds, two compounds were found active, namely, F0539-1482 and F0514-4011. F0514-4011 was the most efficient and impaired this scattering response to HGF with an IC 50 of 7.2 μ M (Figure 4). No toxic effects were observed at biologically active concentrations. This study thus demonstrates that our strategy actually allows the identification of compounds able to inhibit MET-driven biological activities.

3.4. Compared Docking of F0514-4011 Compound Versus Known Inhibitors

In order to understand why the compound F0514-4011 (Figure 5) was the most potent compound among the 25 experimentally tested ones while not presenting the highest GOLD score, we compared its docking data with those of potent existing inhibitors. For that, we collected the structures of ligands found in the PDB related to MET kinase domain in complex with already marketed inhibitors with binding IC 50 found in the nM range (Table 7). All these compounds were submitted to the ensemble-docking GOLD protocol already used for our virtual screening campaign. From these calculations, it appeared that the best docking scores ranged from 111 for merestinib (L1X ID in PDB 4EEV) to 83 for AMG337 (5T1 ID in PDB 5EYD), so that the score of 103 obtained for our active F0514-4011 compound was in this range of active compounds. Considering now IC 50 , one possibility to explain the higher IC 50 of 7.2 μ M obtained for F0514-4011 (compared to 0.4–14 μ M range found for compounds listed in Table 7) could be its weaker solubility (cLogP of 5.7, greater than that of all compounds listed in Table 7).
Another point concerned the interaction of F0514-4011 with amino acid residues within the protein-binding region. In Table 8, we have listed the protein residues/ligand interactions found from Table 7 PDB complexes, as calculated by the PLIP program. These interactions were compared to the ones obtained for F0514-4011 from its best pose MD_3EFJ in the ensemble MET conformations. From this comparison, it appears that several of the most important amino acid residues found from the PDB protein/ligand analysis were also found for F0514-4011, at the exception of Met 1160 , common to all PDB structures of Table 8, replaced possibly by Met 1131 and Met 1229 in our case. This situation is mostly due to the conformation of the large DFG loop acting as a highly flexible lid protecting the binding sites which was quite different in the MD_3EFJ conformation, found as the most suitable one to bind F0514-4011 when compared to the PDB ones (see Figure 6 for an example with the 5DG5 and 4DEI structures). Therefore, our docking results concerning the best pose proposed by GOLD for F0514-4011 appear quite in agreement with most of data obtained from all the PDB concerning MET kinase domain complexed with inhibitors.
To further characterize the F0514-4011 inhibitor type, we have considered the general 3D shape of known kinase inhibitors as analyzed in several papers [7,65,66,67]. Concerning MET, such compounds are generally classified as type-I or -II. Type-I ligands essentially bind at the ATP binding site and present a U-shaped conformation, with the protein in the DFG-in structure; while type II are found in an extended shape and correspond to the DFG-out protein form. We illustrate this in Figure 7, showing the conformations of two typical ligands, namely, type-I AMG337 (from PDB 5EYD) and the type-II altiratinin analog DP-4157 (from PDB 5DG5). From this picture, it appears that F0514-4011 presents both the U and linear shapes while also showing another region of interaction, including three of the protein residues already found in MET complexes—namely, Asp 1222 , Tyr 1230 , and Arg 1208 (found only 2 times for 3C1X and 3YW8 among our 45 ensemble conformations). Asp 1204 and Asn 1209 residues, still not involved in MET complex PDB structures, complement this supplementary binding pocket. The thiophene moiety of F0514-4001 was placed central within this pocket by the thiophene-pyrazole group which also oriented the associated toluene ring to close the U-shape part. Therefore, one could postulate that F0514-4011 molecule describes a possibly novel type of inhibitor.
Nevertheless, considering the limitations of any docking program, the stability of F0514-4011’s best docking pose could be questioned. In order to validate it, we have performed a molecular dynamics simulation using the same conditions as those used for the PDB complexes (cubic water box of 80 Å3). The results show that the GOLD docking pose is very stable and still conserved after 10 ns of MD (Figure 8). The protein/ligand interactions found for F0514-4011 after the MD simulation were similar to those discussed above, thus giving confidence to the robustness of the docking results. Our final question concerns the originality of F0514-4011 compared to the known MET ligands. The Tanimoto similarity index calculated between F0514-4011 and most of the published MET ligands shows that the molecule identified by our virtual screening campaign seems to be an innovative hit as all the Tanimoto values are low, ranging from 0.39 (with the pioneer inhibitor PHA-665752) to 0.12 (for norcantharidin) (Table S2). We have completed this quite elementary similarity search by using the ChemDes web server [68], which allows a large panel of similarity fingerprint types as well as fingerprints descriptors and similarity measures. Using this web server, we mined several databases collecting MET known inhibitors (such as the PDB or PubChem [69]), already in clinical trials (such as MDDR [70]), or described as putative inhibitors (such as in Life Chemical or sellekchem [71] providers). The results obtained with this method confirmed the lack of similarity suggested with the Tanimoto distance. With the Sokal similarity method and DTRF fingerprint types, the similarities ranged from 0.46 to 0.19 (in the PDB list, a maximum of 0.40 was obtained for compound ID 75H found in PDB ID 5T3Q (data not shown)).This could be due to the thiophene moiety of F0514-4011, as we have found only two papers and one patent in the literature referring to thiophene-related MET inhibitors [72,73,74] and only one reference to the role of thiophene-pyrazole moiety in kinase inhibition [75].

4. Discussion

Molecular docking, molecular dynamics, and virtual screening approaches can now be efficiently used for the design of new inhibitors of the MET kinase domain [27,56,76,77,78,79,80]. From all these approaches, new potent compounds were obtained and more highlights revealed about MET kinase domain conformational behavior. In this vein, our study merges both simulations and experiments and highlights a novel scaffold for MET inhibition.
Using an ensemble-docking approach associated to short molecular dynamics runs in order to take into account the flexibility of the used X-ray structures in the protein conformational ensemble, we were faced with the fundamental question of the relevance of this strategy for handling the difficult problem of predicting ligand-binding modes on a flexible target. This is especially true for MET kinase, the active site of which exhibits important structural variations, as observed in their available crystal structures [81,82]. We believe that this work brings a positive answer to this question and can constitute a working line for other simulations in the future. Ensemble-docking is now widely used, and incorporating this approach to short molecular dynamics simulations looks promising. Still, a couple of simple questions have to be answered prior to initiating the docking calculation: how do we generate a relevant ensemble for a given receptor [51], and how can we be sure that the possible energy differences obtained between conformations in the ensemble are properly accounted for?
Interestingly, F0514-4011 compound (also referenced in PubChem with ID 5237313) is not a newcomer in drug design as it has been already tested as a possible activator of E3 ligase (FBW7) and inhibitor of microphthalmia-associated transcription factor (MITF), but was found inactive in both assays. Our study suggests that it could be repositioned for MET inhibition, as evidenced by its biological activity against MET-driven cell scattering. Some drug properties such as solubility and lack of toxicity were already known. With regard to its molecular weight of 650Da, which could be considered as limiting its possible therapeutic action, it should be noted that other inhibitors currently on the market have similar characteristics such as tarloxatinib (679Da), foretinib (632Da), or golvatinib (633Da). Therefore, it should not be a major hurdle if lead optimization provides us with a promising drug in terms of activity and/or selectivity. This will be the topic of future investigations.
This virtual screening work presents F0414-4011 as a valuable compound that could be a seed for developing new and innovative leads against MET kinase. Its novelty and originality might be used to overcome the resistance problem found presently for several existing inhibitors.

Supplementary Materials

The following are available online. Table S1: Comparison of the ensemble-docking results to the individual ones (a ligand against its own PDB-related structure), Table S2: most used c-Met inhibitors as pointed by SelleckChem and AdooQ Biosciences.

Author Contributions

Conceptualization, A.F., V.L., and B.M.; software, E.B. and P.N.; validation, A.F., F.M., and R.D.; formal analysis, E.B., A.F., P.N., and B.M.; investigation, E.B, P.N., V.L., and B.M.; writing–original draft preparation, E.B., A.F., P.N., V.L., and B.M.; writing–review and editing, E.B., A.F., P.N., and B.M.; supervision, F.M., R.D., and B.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Experiments presented in this paper were carried out using the Grid’5000 testbed, supported by a scientific interest group hosted by Inria and including CNRS, RENATER, and several Universities as well as other organizations (see https://www.grid5000.fr). We are much grateful to all members of our labs for helpful discussion and advice.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
DFGaspartate-phenylalanine-glycine
MDMolecular dynamics
RMSDRoot mean square deviation

References

  1. Furlan, A.; Kherrouche, Z.; Montagne, R.; Copin, M.C.; Tulasne, D. Thirty Years of Research on Met Receptor to Move a Biomarker from Bench to Bedside. Cancer Res. 2014, 74, 6737–6744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zhang, H.; Feng, Q.; Chen, W.D.; Wang, Y.D. HGF/c-MET: A Promising Therapeutic Target in the Digestive System Cancers. Int. J. Mol. Sci. 2018, 19, 3295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Rashed, W.M. C-MET as a Potential Target Therapy toward Personalized Therapy in Some Pediatric Tumors: An Overview. Crit. Rev. Oncol. 2018, 131, 7–15. [Google Scholar] [CrossRef] [PubMed]
  4. Bahrami, A.; Shahidsales, S.; Khazaei, M.; Ghayour-Mobarhan, M.; Maftouh, M.; Hassanian, S.M.; Avan, A. C-Met as a Potential Target for the Treatment of Gastrointestinal Cancer: Current Status and Future Perspectives. J. Cell. Physiol. 2017, 232, 2657–2673. [Google Scholar] [CrossRef] [PubMed]
  5. Refaat, T.; Donnelly, E.D.; Sachdev, S.; Parimi, V.; El Achy, S.; Dalal, P.; Farouk, M.; Berg, K.N.; Helenowksi, I.; Gross, J.P.; et al. C-Met Overexpression in Cervical Cancer, a Prognostic Factor and a Potential Molecular Therapeutic Target. Am. J. Clin. Oncol. 2017, 40, 590. [Google Scholar] [CrossRef] [PubMed]
  6. Trovato, M.; Campennì, A.; Giovinazzo, S.; Siracusa, M.; Ruggeri, R.M. Hepatocyte Growth Factor/C-Met Axis in Thyroid Cancer: From Diagnostic Biomarker to Therapeutic Target. Biomark. Insights 2017, 12, 1177271917701126. [Google Scholar] [CrossRef] [Green Version]
  7. Wu, P.; Clausen, M.H.; Nielsen, T.E. Allosteric small-molecule kinase inhibitors. Pharmacol. Ther. 2015, 156, 59–68. [Google Scholar] [CrossRef] [Green Version]
  8. Mo, H.N.; Liu, P. Targeting MET in Cancer Therapy. Chronic Dis. Transl. Med. 2017, 3, 148–153. [Google Scholar] [CrossRef]
  9. Li, C.; Wu, J.; Hynes, M.; Dosch, J.; Sarkar, B.; Welling, T.H.; di Magliano, M.P.; Simeone, D.M. C-Met Is a Marker of Pancreatic Cancer Stem Cells and Therapeutic Target. Gastroenterology 2011, 141, 2218–2227. [Google Scholar] [CrossRef]
  10. Sawada, K.; Radjabi, A.R.; Shinomiya, N.; Kistner, E.; Kenny, H.; Becker, A.R.; Turkyilmaz, M.A.; Salgia, R.; Yamada, S.D.; Woude, G.F.V.; et al. C-Met Overexpression Is a Prognostic Factor in Ovarian Cancer and an Effective Target for Inhibition of Peritoneal Dissemination and Invasion. Cancer Res. 2007, 67, 1670–1679. [Google Scholar] [CrossRef] [Green Version]
  11. Sierra, J.R.; Tsao, M.S. C-MET as a Potential Therapeutic Target and Biomarker in Cancer. Ther. Adv. Med. Oncol. 2011, 3, S21–S35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Cui, J.J. Targeting Receptor Tyrosine Kinase MET in Cancer: Small Molecule Inhibitors and Clinical Progress. J. Med. Chem. 2014, 57, 4427–4453. [Google Scholar] [CrossRef] [PubMed]
  13. Parikh, P.K.; Ghate, M.D. Recent Advances in the Discovery of Small Molecule C-Met Kinase Inhibitors. Eur. J. Med. Chem. 2018, 143, 1103–1138. [Google Scholar] [CrossRef] [PubMed]
  14. Pasquini, G.; Giaccone, G. C-MET Inhibitors for Advanced Non-Small Cell Lung Cancer. Expert Opin. Investig. Drugs 2018, 27, 363–375. [Google Scholar] [CrossRef] [PubMed]
  15. Yuan, H.; Liu, Q.; Zhang, L.; Hu, S.; Chen, T.; Li, H.; Chen, Y.; Xu, Y.; Lu, T. Discovery, Optimization and Biological Evaluation for Novel c-Met Kinase Inhibitors. Eur. J. Med. Chem. 2018, 143, 491–502. [Google Scholar] [CrossRef] [PubMed]
  16. Zhu, K.; Kong, X.; Zhao, D.; Liang, Z.; Luo, C. C-MET Kinase Inhibitors: A Patent Review (2011–2013). Expert Opin. Ther. Patents 2014, 24, 217–230. [Google Scholar] [CrossRef] [PubMed]
  17. Lv, P.C.; Wang, Z.C.; Zhu, H.L. Recent Advances in the Design and Synthesis of C-Met Inhibitors as Anticancer Agents (2014-Present). Curr. Med. Chem. 2017, 24, 57–64. [Google Scholar] [CrossRef]
  18. Sun, Z.G.; Yang, Y.A.; Zhang, Z.G.; Zhu, H.L. Optimization techniques for novel c-Met kinase inhibitors. Expert Opin. Drug Discov. 2019, 14, 59–69. [Google Scholar] [CrossRef]
  19. Hughes, V.S.; Siemann, D.W. Have Clinical Trials Properly Assessed C-Met Inhibitors? Trends Cancer 2018, 4, 94–97. [Google Scholar] [CrossRef]
  20. Miranda, O.; Farooqui, M.; Siegfried, J. Status of Agents Targeting the HGF/c-Met Axis in Lung Cancer. Cancers 2018, 10, 280. [Google Scholar] [CrossRef] [Green Version]
  21. Cortot, A.B.; Kherrouche, Z.; Descarpentries, C.; Wislez, M.; Baldacci, S.; Furlan, A.; Tulasne, D. Exon 14 deleted MET receptor as a new biomarker and target in cancers. JNCI J. Natl. Cancer Inst. 2017, 109, djw262. [Google Scholar] [CrossRef] [PubMed]
  22. Asses, Y.; Venkatraman, V.; Leroux, V.; Ritchie, D.W.; Maigret, B. Exploring C-Met Kinase Flexibility by Sampling and Clustering Its Conformational Space. Proteins Struct. Funct. Bioinform. 2012, 80, 1227–1238. [Google Scholar] [CrossRef] [PubMed]
  23. Dussault, I.; Bellon, S.F. C-Met Inhibitors with Different Binding Modes: Two Is Better than One. Cell Cycle 2008, 7, 1157–1160. [Google Scholar] [CrossRef] [PubMed]
  24. Jacobs, M.D.; Caron, P.R.; Hare, B.J. Classifying Protein Kinase Structures Guides Use of Ligand-Selectivity Profiles to Predict Inactive Conformations: Structure of Lck/Imatinib Complex. Proteins Struct. Funct. Bioinform. 2008, 70, 1451–1460. [Google Scholar] [CrossRef]
  25. Druker, B.J.; Talpaz, M.; Resta, D.J.; Peng, B.; Buchdunger, E.; Ford, J.M.; Lydon, N.B.; Kantarjian, H.; Capdeville, R.; Ohno-Jones, S.; et al. Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. N. Engl. J. Med. 2001, 344, 1031–1037. [Google Scholar] [CrossRef] [Green Version]
  26. Capdeville, R.; Buchdunger, E.; Zimmermann, J.; Matter, A. Glivec (STI571, Imatinib), a Rationally Developed, Targeted Anticancer Drug. Nat. Rev. Drug Discov. 2002, 1, 493. [Google Scholar] [CrossRef]
  27. Aliebrahimi, S.; Kouhsari, S.M.; Ostad, S.N.; Arab, S.S.; Karami, L. Identification of Phytochemicals Targeting C-Met Kinase Domain Using Consensus Docking and Molecular Dynamics Simulation Studies. Cell Biochem. Biophys. 2018, 76, 135–145. [Google Scholar] [CrossRef]
  28. Li, M.J.; Wu, G.Z.; Kaas, Q.; Jiang, T.; Yu, R.L. Development of Efficient Docking Strategies and Structure-Activity Relationship Study of the c-Met Type II Inhibitors. J. Mol. Graph. Model. 2017, 75, 241–249. [Google Scholar] [CrossRef] [Green Version]
  29. Amaro, R.E.; Baudry, J.; Chodera, J.; Demir, O.; McCammon, J.A.; Miao, Y.; Smith, J.C. Ensemble Docking in Drug Discovery. Biophys. J. 2018, 114, 2271–2278. [Google Scholar] [CrossRef]
  30. Patané, S.; Pietrancosta, N.; Hassani, H.; Leroux, V.; Maigret, B.; Kraus, J.L.; Dono, R.; Maina, F. A New Met Inhibitory-Scaffold Identified by a Focused Forward Chemical Biological Screen. Biochem. Biophys. Res. Commun. 2008, 375, 184–189. [Google Scholar] [CrossRef]
  31. Furlan, A.; Colombo, F.; Kover, A.; Issaly, N.; Tintori, C.; Angeli, L.; Leroux, V.; Letard, S.; Amat, M.; Asses, Y.; et al. Identification of New Aminoacid Amides Containing the Imidazo [2, 1-b] Benzothiazol-2-Ylphenyl Moiety as Inhibitors of Tumorigenesis by Oncogenic Met Signaling. Eur. J. Med. Chem. 2012, 47, 239–254. [Google Scholar] [CrossRef] [PubMed]
  32. Asses, Y.; Leroux, V.; Tairi-Kellou, S.; Dono, R.; Maina, F.; Maigret, B. Analysis of C-Met Kinase Domain Complexes: A New Specific Catalytic Site Receptor Model for Defining Binding Modes of ATP-Competitive Ligands. Chem. Biol. Drug Des. 2009, 74, 560–570. [Google Scholar] [CrossRef] [PubMed]
  33. Gimeno, A.; Ojeda-Montes, M.; Tomás-Hernández, S.; Cereto-Massagué, A.; Beltrán-Debón, R.; Mulero, M.; Pujadas, G.; Garcia-Vallvé, S. The Light and Dark Sides of Virtual Screening: What Is There to Know? Int. J. Mol. Sci. 2019, 20, 1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kioshima, E.S.; Shinobu-Mesquita, C.S.; Abadio, A.K.R.; Felipe, M.S.S.; Svidzinski, T.I.E.; Maigret, B. Selection of potential anti-adhesion drugs by in silico approaches targeted to ALS3 from Candida albicans. Biotechnol. Lett. 2019, 41, 1391–1401. [Google Scholar] [CrossRef]
  35. Bresso, E.; Fernandez, D.; Amora, D.X.; Noel, P.; Petitot, A.S.; de Sa, M.E.L.; Albuquerque, E.V.S.; Danchin, E.G.J.; Maigret, B.; Martins, N.F. A Chemosensory GPCR as a Potential Target to Control the Root-Knot Nematode Meloidogyne incognita Parasitism in Plants. Molecules 2019, 24, 3798. [Google Scholar] [CrossRef] [Green Version]
  36. Rodrigues-Vendramini, F.A.V.; Faria, D.R.; Arita, G.S.; Capoci, I.R.G.; Sakita, K.M.; Caparroz-Assef, S.M.; Becker, T.C.A.; de Souza Bonfim-Mendonça, P.; Felipe, M.S.; Svidzinski, T.I.E.; et al. Antifungal activity of two oxadiazole compounds for the paracoccidioidomycosis treatment. PLoS Negl. Trop. Dis. 2019, 13, e0007441. [Google Scholar] [CrossRef] [Green Version]
  37. Gilad, Y.; Nadassy, K.; Senderowitz, H. A Reliable Computational Workflow for the Selection of Optimal Screening Libraries. J. Cheminform. 2015, 7, 61. [Google Scholar] [CrossRef] [Green Version]
  38. Petrone, P.M.; Wassermann, A.M.; Lounkine, E.; Kutchukian, P.; Simms, B.; Jenkins, J.; Selzer, P.; Glick, M. Biodiversity of Small Molecules–a New Perspective in Screening Set Selection. Drug Discov. Today 2013, 18, 674–680. [Google Scholar] [CrossRef]
  39. Huggins, D.J.; Venkitaraman, A.R.; Spring, D.R. Rational Methods for the Selection of Diverse Screening Compounds. ACS Chem. Biol. 2011, 6, 208–217. [Google Scholar] [CrossRef] [Green Version]
  40. Ma, C.; Lazo, J.S.; Xie, X.Q. Compound Acquisition and Prioritization Algorithm for Constructing Structurally Diverse Compound Libraries. ACS Comb. Sci. 2011, 13, 223–231. [Google Scholar] [CrossRef] [Green Version]
  41. Xi, H.; Lunney, E.A. The Design, Annotation, and Application of a Kinase-Targeted Library. In Chemical Library Design; Humana Press: Totowa, NJ, USA, 2011; pp. 279–291. [Google Scholar]
  42. Deanda, F.; Stewart, E.L.; Reno, M.J.; Drewry, D.H. Kinase-Targeted Library Design through the Application of the PharmPrint Methodology. J. Chem. Inf. Model. 2008, 48, 2395–2403. [Google Scholar] [CrossRef] [PubMed]
  43. Dutta, S.; Mahalanobish, S.; Saha, S.; Ghosh, S.; Sil, P.C. Natural Products: An Upcoming Therapeutic Approach to Cancer. Food Chem. Toxicol. 2019, 128, 240–255. [Google Scholar] [CrossRef] [PubMed]
  44. Henkin, J.M.; Ren, Y.; Soejarto, D.D.; Kinghorn, A.D. The Search for Anticancer Agents from Tropical Plants. In Progress in the Chemistry of Organic Natural Products 107; Springer: New York, NY, USA, 2018; pp. 1–94. [Google Scholar]
  45. Mondal, S.; Bandyopadhyay, S.; K Ghosh, M.; Mukhopadhyay, S.; Roy, S.; Mandal, C. Natural Products: Promising Resources for Cancer Drug Discovery. Anti-Cancer Agents Med. Chem. (Former Curr. Med.-Chem.-Anti-Cancer Agents) 2012, 12, 49–75. [Google Scholar] [CrossRef]
  46. Nowak-Sliwinska, P.; Scapozza, L.; i Altaba, A.R. Drug Repurposing in Oncology: Compounds, Pathways, Phenotypes and Computational Approaches for Colorectal Cancer. Biochim. Biophys. Acta (BBA) Rev. Cancer 2019, 1871, 434–454. [Google Scholar] [CrossRef] [PubMed]
  47. Cheng, F. In Silico Oncology Drug Repositioning and Polypharmacology. In Cancer Bioinformatics; Springer: New York, NY, USA, 2019; pp. 243–261. [Google Scholar]
  48. Abdelaleem, M.; Ezzat, H.; Osama, M.; Megahed, A.; Alaa, W.; Gaber, A.; Shafei, A.; Refaat, A. Prospects for Repurposing CNS Drugs for Cancer Treatment. Oncol. Rev. 2019, 13, 411. [Google Scholar] [CrossRef] [PubMed]
  49. Yadav, V.; Talwar, P. Repositioning of Fluoroquinolones from Antibiotic to Anti-Cancer Agents: An Underestimated Truth. Biomed. Pharmacother. 2019, 111, 934–946. [Google Scholar] [CrossRef]
  50. Lipinski, C.A. Lead-and Drug-like Compounds: The Rule-of-Five Revolution. Drug Discov. Today Technol. 2004, 1, 337–341. [Google Scholar] [CrossRef]
  51. Evangelista Falcon, W.; Ellingson, S.R.; Smith, J.C.; Baudry, J. Ensemble Docking in Drug Discovery: How Many Protein Configurations from Molecular Dynamics Simulations are Needed To Reproduce Known Ligand Binding? J. Phys. Chem. B 2019, 123, 5189–5195. [Google Scholar] [CrossRef]
  52. Motta, S.; Bonati, L. Modeling Binding with Large Conformational Changes: Key Points in Ensemble-Docking Approaches. J. Chem. Inf. Model. 2017, 57, 1563–1578. [Google Scholar] [CrossRef]
  53. Berman, H.M. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef] [Green Version]
  54. Rahman, R.; Ung, P.M.U.; Schlessinger, A. KinaMetrix: A web resource to investigate kinase conformations and inhibitor space. Nucleic Acids Res. 2019, 47, D361–D366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Holm, L.; Laakso, L.M. Dali server update. Nucleic Acids Res. 2016, 44, W351–W355. [Google Scholar] [CrossRef] [PubMed]
  56. Ibrahim, H.S.; Albakri, M.E.; Mahmoud, W.R.; Allam, H.A.; Reda, A.M.; Abdel-Aziz, H.A. Synthesis and Biological Evaluation of Some Novel Thiobenzimidazole Derivatives as Anti-Renal Cancer Agents through Inhibition of c-MET Kinase. Bioorg. Chem. 2019, 85, 337–348. [Google Scholar] [CrossRef] [PubMed]
  57. Phillips, J.C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R.D.; Kalé, L.; Schulten, K. Scalable Molecular Dynamics with NAMD. J. Comput. Chem. 2005, 26, 1781–1802. [Google Scholar] [CrossRef] [Green Version]
  58. Jones, G.; Willett, P.; Glen, R.C.; Leach, A.R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking. J. Mol. Biol. 1997, 267, 727–748. [Google Scholar] [CrossRef] [Green Version]
  59. Hendlich, M.; Rippmann, F.; Barnickel, G. LIGSITE: Automatic and Efficient Detection of Potential Small Molecule-Binding Sites in Proteins. J. Mol. Graph. Model. 1997, 15, 359–363. [Google Scholar] [CrossRef]
  60. Bolze, R.; Cappello, F.; Caron, E.; Daydé, M.; Desprez, F.; Jeannot, E.; Jégou, Y.; Lanteri, S.; Leduc, J.; Melab, N.; et al. Grid’5000: A Large Scale and Highly Reconfigurable Experimental Grid Testbed. Int. J. High Perform. Comput. Appl. 2006, 20, 481–494. [Google Scholar] [CrossRef]
  61. Ghemtio, L.; Jeannot, E.; Maigret, B. Efficiency of a Hierarchical Protocol for High Throughput Structure-Based Virtual Screening on GRID5000 Cluster Grid. Open Access Bioinform. 2010, 2, 41–53. [Google Scholar]
  62. Furlan, A.; Roux, B.; Lamballe, F.; Conti, F.; Issaly, N.; Daian, F.; Guillemot, J.F.; Richelme, S.; Contensin, M.; Bosch, J.; et al. Combined Drug Action of 2-Phenylimidazo [2, 1-b] Benzothiazole Derivatives on Cancer Cells According to Their Oncogenic Molecular Signatures. PLoS ONE 2012, 7, e46738. [Google Scholar] [CrossRef] [Green Version]
  63. Salentin, S.; Schreiber, S.; Haupt, V.J.; Adasme, M.F.; Schroeder, M. PLIP: Fully Automated Protein–Ligand Interaction Profiler. Nucleic Acids Res. 2015, 43, W443–W447. [Google Scholar] [CrossRef]
  64. Colombo, F.; Tintori, C.; Furlan, A.; Borrelli, S.; Christodoulou, M.S.; Dono, R.; Maina, F.; Botta, M.; Amat, M.; Bosch, J.; et al. ‘Click’ synthesis of a triazole-based inhibitor of Met functions in cancer cells. Bioorg. Med. Chem. Lett. 2012, 22, 4693–4696. [Google Scholar] [CrossRef] [PubMed]
  65. Roskoski, R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol. Res. 2016, 103, 26–48. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, Z.; Xie, L.; Bourne, P.E. Structural Insights into Characterizing Binding Sites in Epidermal Growth Factor Receptor Kinase Mutants. J. Chem. Inf. Model. 2019, 59, 453–462. [Google Scholar] [CrossRef] [PubMed]
  67. Panicker, R.C.; Chattopadhaya, S.; Coyne, A.G.; Srinivasan, R. Allosteric Small-Molecule Serine/Threonine Kinase Inhibitors. In Protein Allostery in Drug Discovery; Springer: Singapore, 2019; Volume 1163, pp. 253–278. [Google Scholar]
  68. Dong, J.; Cao, D.S.; Miao, H.Y.; Liu, S.; Deng, B.C.; Yun, Y.H.; Wang, N.N.; Lu, A.P.; Zeng, W.B.; Chen, A.F. ChemDes: An integrated web-based platform for molecular descriptor and fingerprint computation. J. Cheminform. 2015, 7, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Kim, S.; Thiessen, P.A.; Bolton, E.E.; Chen, J.; Fu, G.; Gindulyte, A.; Han, L.; He, J.; He, S.; Shoemaker, B.A.; et al. PubChem Substance and Compound databases. Nucleic Acids Res. 2016, 44, D1202–D1213. [Google Scholar] [CrossRef] [PubMed]
  70. MDDR. Available online: www.3dsbiovia.com/products/collaborative-science/databases/bioactivity-databases/mddr.html (accessed on 18 February 2020).
  71. Selleckchem.Com-Inhibitor Expert. Available online: www.selleckchem.com (accessed on 18 February 2020).
  72. Mohareb, R.M.; Hilmy, K.M.; Elshehawy, Y.A. Discovery of new thiophene, pyrazole, isoxazole derivatives as antitumor, c-Met, tyrosine kinase and Pim-1 kinase inhibitors. Bull. Chem. Soc. Ethiop. 2018, 32, 285. [Google Scholar] [CrossRef] [Green Version]
  73. Shi, L.; Wu, T.T.; Wang, Z.; Xue, J.Y.; Xu, Y.G. Discovery of quinazolin-4-amines bearing benzimidazole fragments as dual inhibitors of c-Met and VEGFR-2. Bioorg. Med. Chem. 2014, 22, 4735–4744. [Google Scholar] [CrossRef]
  74. Fancelli, D.; Pevarello, P.; Varasi, M. Thiophene Derivatives Active as Kinase Inhibitors, Process for Their Preparation and Pharmaceutical Compositions Comprising Them. WO2001EP06763, 14 June 2001. [Google Scholar]
  75. Zhan, W.; Che, J.; Xu, L.; Wu, Y.; Hu, X.; Zhou, Y.; Cheng, G.; Hu, Y.; Dong, X.; Li, J. Discovery of pyrazole-thiophene derivatives as highly Potent, orally active Akt inhibitors. Eur. J. Med. Chem. 2019, 180, 72–85. [Google Scholar] [CrossRef]
  76. Zhang, S.; Song, Q.; Wang, X.; Wei, Z.; Yu, R.; Wang, X.; Jiang, T. Virtual Screening Guided Design, Synthesis and Bioactivity Study of Benzisoselenazolones (BISAs) on Inhibition of c-Met and Its Downstream Signalling Pathways. Int. J. Mol. Sci. 2019, 20, 2489. [Google Scholar] [CrossRef] [Green Version]
  77. Balasubramanian, P.K.; Balupuri, A.; Bhujbal, S.P.; Cho, S.J. 3D-QSAR-Aided Design of Potent c-Met Inhibitors Using Molecular Dynamics Simulation and Binding Free Energy Calculation. J. Biomol. Struct. Dyn. 2019, 37, 2165–2178. [Google Scholar] [CrossRef]
  78. Zhang, Q.W.; Ye, Z.D.; Shen, C.; Tie, H.X.; Wang, L.; Shi, L. Synthesis of Novel 6, 7-Dimethoxy-4-Anilinoquinolines as Potent c-Met Inhibitors. J. Enzym. Inhib. Med. Chem. 2019, 34, 124–133. [Google Scholar] [CrossRef] [PubMed]
  79. Singh, P.K.; Silakari, O. Molecular Dynamics Guided Development of Indole Based Dual Inhibitors of EGFR (T790M) and c-MET. Bioorg. Chem. 2018, 79, 163–170. [Google Scholar] [CrossRef] [PubMed]
  80. Yan, M.; Wang, H.; Wang, Q.; Zhang, Z.; Zhang, C. Allosteric inhibition of c-Met kinase in sub-microsecond molecular dynamics simulations induced by its inhibitor, tivantinib. Phys. Chem. Chem. Phys. 2016, 18, 10367–10374. [Google Scholar] [CrossRef] [PubMed]
  81. Modi, V.; Dunbrack, R.L. Defining a new nomenclature for the structures of active and inactive kinases. Proc. Natl. Acad. Sci. USA 2019, 116, 6818–6827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mashayekh, K.; Sharifi, S.; Damghani, T.; Elyasi, M.; Avestan, M.S.; Pirhadi, S. Clustering and Sampling of the c-Met Conformational Space: A Computational Drug Discovery Study. Comb. Chem. High Throughput Screen. 2020, 22, 635–648. [Google Scholar] [CrossRef]
Figure 1. (a) Dendrogram showing the relationships between the 45 PDB conformers listed in Table 2 and Table 3 and used to sample MET structure plasticity. (b) Similarity heat-map showing the relationships between the 45 PDB conformers and used to sample MET structure plasticity. The color scale corresponds to the Dali Z-score values. (c) Correspondence analysis of the 45 ensemble PDB-related conformers. This plot positions data points with the most similar structural neighborhoods near each other according to a multidimensional scaling method.
Figure 1. (a) Dendrogram showing the relationships between the 45 PDB conformers listed in Table 2 and Table 3 and used to sample MET structure plasticity. (b) Similarity heat-map showing the relationships between the 45 PDB conformers and used to sample MET structure plasticity. The color scale corresponds to the Dali Z-score values. (c) Correspondence analysis of the 45 ensemble PDB-related conformers. This plot positions data points with the most similar structural neighborhoods near each other according to a multidimensional scaling method.
Molecules 25 00938 g001
Figure 2. Position of the average center-point (as a green sphere) found from the 45 used conformers and used for the ensemble-dockings.
Figure 2. Position of the average center-point (as a green sphere) found from the 45 used conformers and used for the ensemble-dockings.
Molecules 25 00938 g002
Figure 3. Poses of the AM7 ligand in the X-ray 2RFN structure compared to the docking results. In black, the original pose of the ligand in its PDB protein conformation; in colors, the best docking poses obtained by GOLD on the 2RFN target using a definition of the binding site from a list of residues (orange), from the original ligand (green), and from a center-point (purple).
Figure 3. Poses of the AM7 ligand in the X-ray 2RFN structure compared to the docking results. In black, the original pose of the ligand in its PDB protein conformation; in colors, the best docking poses obtained by GOLD on the 2RFN target using a definition of the binding site from a list of residues (orange), from the original ligand (green), and from a center-point (purple).
Molecules 25 00938 g003
Figure 4. (a) F0514-4011 impairs cell scattering in response to MET ligand HGF: MDCK epithelial cells were treated with 20 ng/mL HGF, with or without preincubation with F0514-4011 for 2 h. F0514-4011 IC 50 is 7.2 μ M. (b) Dose-response curve for F0514-4011.
Figure 4. (a) F0514-4011 impairs cell scattering in response to MET ligand HGF: MDCK epithelial cells were treated with 20 ng/mL HGF, with or without preincubation with F0514-4011 for 2 h. F0514-4011 IC 50 is 7.2 μ M. (b) Dose-response curve for F0514-4011.
Molecules 25 00938 g004
Figure 5. F0514-4011: N-[[4-(4-Ethoxyphenyl)-5-[2-[3-(4-methylphenyl)-5-thiophen-2-yl-3,4-dihydropyrazol -2-yl]-2-oxoethyl]sulfanyl-1,2,4-triazol-3-yl]methyl]-2-phenylacetamide.
Figure 5. F0514-4011: N-[[4-(4-Ethoxyphenyl)-5-[2-[3-(4-methylphenyl)-5-thiophen-2-yl-3,4-dihydropyrazol -2-yl]-2-oxoethyl]sulfanyl-1,2,4-triazol-3-yl]methyl]-2-phenylacetamide.
Molecules 25 00938 g005
Figure 6. Differences for the lid DFG loop between selected PDB structures and our MD-refined MD_3EFJ conformations. The proteins are depicted from their C α ribbon-like traces.
Figure 6. Differences for the lid DFG loop between selected PDB structures and our MD-refined MD_3EFJ conformations. The proteins are depicted from their C α ribbon-like traces.
Molecules 25 00938 g006
Figure 7. Comparison of the conformations between F0514-4011, the U-shape inhibitor 5T1 (AMG337), and the linear-shape 5B4 (altiratinib), as observed in their respective binding sites.
Figure 7. Comparison of the conformations between F0514-4011, the U-shape inhibitor 5T1 (AMG337), and the linear-shape 5B4 (altiratinib), as observed in their respective binding sites.
Molecules 25 00938 g007
Figure 8. (a) Comparison between the initial docking pose (in orange) of F0514-4011 and the final one after the 10-ns MD simulation (in cyan). (b) Evolution of F0514-4011 root mean square deviation (RMSD) during 11 ns (1 ns equilibrium and 10 ns production) of MD simulation. Poses were aligned on the initial one and the curve was smoothed.
Figure 8. (a) Comparison between the initial docking pose (in orange) of F0514-4011 and the final one after the 10-ns MD simulation (in cyan). (b) Evolution of F0514-4011 root mean square deviation (RMSD) during 11 ns (1 ns equilibrium and 10 ns production) of MD simulation. Poses were aligned on the initial one and the curve was smoothed.
Molecules 25 00938 g008
Table 1. List of the selected chemical libraries used in the present virtual screening campaign, providing a total of 76,251 compounds.
Table 1. List of the selected chemical libraries used in the present virtual screening campaign, providing a total of 76,251 compounds.
SupplierLibrary Names
French laboratories chimiotheque-nationale.enscm.frSelected subsets (kinase, essential, etc.)
Collaboration medchem.u-strasbg.frlaboratory collection
ChemBridge www.chembridge.com/index.phpkinase library diversity library
Life Chemicals www.lifechemicals.comkinase general library antiviral library
15K diversity library
TimTec www.timtec.netNDL + NPL natural derivatives library
Otava otavachemicals.comall kinase library
10K diversity library
natural productlike library
TargetMol www.targetmol.comantivirus library natural compounds library
Table 2. List of the 26 PDB MET kinase domain structures selected in the previous work [22] and reused in the present one. The kinase conformation associated to each structure is annotated according to the KinaMetrix web resource [56]: DO means DFG-out, DI means DFG-in, CO means α -cHelix-out, CI means α -cHelix-in, and ω CD indicates DFG intermediate.
Table 2. List of the 26 PDB MET kinase domain structures selected in the previous work [22] and reused in the present one. The kinase conformation associated to each structure is annotated according to the KinaMetrix web resource [56]: DO means DFG-out, DI means DFG-in, CO means α -cHelix-out, CI means α -cHelix-in, and ω CD indicates DFG intermediate.
NoPDB IDLigand PDB IDDeposition DateAnnotation
11R0PKSA2003Inactive CODI
22RFNAM72007?
32RFSAM82007Inactive CODI
43C1XCKK2008Inactive CIDO
53CCNLKG2008Inactive CODI
63CD8L5G2008Inactive CODI
73CE31FN2008Inactive CODO
83CTH3192008Inactive CIDO
93CTJ3202008Inactive CIDO
103F66IHX2008Inactive CODI
113F823532008Inactive CIDO
123EFJMT32008Inactive CODO
133EFKMT42008Inactive CODI
142WD1ZZY2009Inactive CODI
153DKCATP2009Inactive CODI
163DKFSX82009Inactive CODI
172WGJVGH2009Active CIDI
183A4PDFQ2010Inactive CODI
193L8VL8V2010Inactive CIDO
203I5NB2D2010Inactive CODI
213LQ888Z2010Inactive CODO
222WKMPFY2010Inactive CODI
233Q6WQ6W2011Active CIDI
243QTI3QT2011Inactive CODI
253RHKM972011Inactive ω CD
263ZXZKRW2011Inactive CODI
Table 3. List of the PDB MET kinase domain structures added to the ones coming from our previous work [22]. The kinase conformation associated to each structure is annotated according to the KinaMetrix web resource [56]: DO means DFG-out, DI means DFG-in, CO means α -cHelix-out, CI means α -cHelix-in, and ω CD indicates DFG intermediate.
Table 3. List of the PDB MET kinase domain structures added to the ones coming from our previous work [22]. The kinase conformation associated to each structure is annotated according to the KinaMetrix web resource [56]: DO means DFG-out, DI means DFG-in, CO means α -cHelix-out, CI means α -cHelix-in, and ω CD indicates DFG intermediate.
NoPDB IDLigand IDLigand IC 50 (nM)DateMissing Sequence# Missing ResiduesAnnotation
14DEI0JL0.6–220121100–1103 1115–11177Inactive CODI
24GG50J30.920121146–11516Inactive CODI
34EEVL1X4.7/4220121225–124319Inactive CIDO
43VW8DF6220131237–1242 1286–129011?
54IWD1JC120131234–1235 1240–12436Active CIDI
63ZCL5TF1920131100–11023Inactive CODI
73ZC5W9Z620131099–11024Inactive CODI
83ZBX6XE520131089–110214Inactive CODI
94KNB1RU47/41020131099–1103 1113–11158Inactive CODI
104MXCDWF6.720141238–12425Inactive CODO
114XYF44X1/520151099–11035
124R1V3E840020151150–11512Inactive CODI
134XMO46G220151098–11036Inactive CODI
145DG55B4?2015--Inactive CODO
155EYD5T1120161098–1103 1151–11528Inactive CODI
165EOB5QQ0.2420161238–12403Inactive CODI
175EYC5SZ320161099–11035Inactive CODI
185UAF84P?20171098–1105 1145–115216Active CIDI
195HTI66L?20171238–12425Inactive CODO
Table 4. List of the secondary structure elements used for aligning all the conformers.
Table 4. List of the secondary structure elements used for aligning all the conformers.
DomainSecondary Structure NameResidues
N-terminal β 11076 to 1081
β 21092 to 1098
β 21104 to 1110
β 21144 to 1146
β 21154 to 1158
C-terminal α E1178 to 1198
α F1263 to 1278
α H1310 to 1320
α I1330 to 1343
Table 5. Comparison of the docking results using the 3 binding site definitions.
Table 5. Comparison of the docking results using the 3 binding site definitions.
Definition of the Binding SiteTarget PDB NameLigand PDB NameRank NumberScore Value
Center + radius 20 Å3DKCATP1105.5
2RFNAM71100.8
Residues list3DKCATP1102.8
2RFNAM7198.0
From its PDB ligand3DKCATP1107.1
2RFNAM71106.6
Table 6. Ligands selected from the Life Chemical (LC) antiviral library and experimentally tested. “-”: the compound (assessed at a concentration up to 100 μ M) did not affect MDCK cell scattering in response to HGF/SF. “+”: the compound impaired MDCK cell scattering in response to HGF/SF with an IC 50 > 10 μ M. “+++”: the compound impaired MDCK cell scattering in response to HGF/SF with an IC 50 < 10 μ M.
Table 6. Ligands selected from the Life Chemical (LC) antiviral library and experimentally tested. “-”: the compound (assessed at a concentration up to 100 μ M) did not affect MDCK cell scattering in response to HGF/SF. “+”: the compound impaired MDCK cell scattering in response to HGF/SF with an IC 50 > 10 μ M. “+++”: the compound impaired MDCK cell scattering in response to HGF/SF with an IC 50 < 10 μ M.
Mol IDLife Chemicals NameGOLD ScoreBest Protein ConformerBiological Activity
1F0725-0356120.7MD_3EFK-
2F0772-0607111.8MD_3EFK-
3F0816-0342111.2MD_3F82-
4F0737-0405110.6MD_3EFJ-
5F0737-0393110.1MD_3EFK-
6F0301-0263105.5MD_3EFK-
7F0721-0868105.1MD_3EFK-
8F0715-0299105.0MD_3EFK-
9F0539-1482104.0MD_3EFJ+
10F0385-0029103.8MD_3EFK-
11F0385-0334103.4MD_3EFJ-
12F0514-4011103.3MD_3EFJ+++
13F0174-0048102.4MD_3EFK-
14F1620-0074102.1MD_3EFJ-
15F0011-0324102.0MD_3EFK-
16F0721-0906102.0MD_3EFJ-
17F0012-0227101.9MD_3EFJ-
18F0721-0911101.9MD_3EFJ-
19F0715-0300101.8MD_2RFN-
20F2252-0240101.1MD_3EFK-
21F0772-2099100.9MD_3EFJ-
22F0473-0261100.9MD_3CE3-
23F0721-0900100.5MD_3EFK-
24F0772-2147100.5MD_3EFJ-
25F0526-0094100.3MD_3EFJ-
Table 7. Data used for some known marketed inhibitors with nano-molar range IC 50 found in the PDB.
Table 7. Data used for some known marketed inhibitors with nano-molar range IC 50 found in the PDB.
PDB IDLigand IDNameIC50Solubility cLogPDocking Score
2RFSAM8SU1127410 nM2.986
2WGJVGHCriotinib11 nM3.582
2WKMPFYPHA-6657529 nM4.488
3DKFSX8SGX-5234 nM1.484
3RHKM97Tivantinib4 nM3.183
3LQ888ZFortinib0.4 nM4.399
3Q6WQ6WMK-24610.4 nM3.393
3QTI3QTNVP-BVU97214 nM1.684
3ZXZKRWPF-042179035 nM0.287
4EEVL1XMerestinib5 nM3.4111
5EYD5T1AMG3371 nM0.383
Table 8. List of the protein residues interacting with a nM. inhibitor from the PDB complexes of Table 7 ranked by their number of occurrence. In bold, the residues also found in the interactions with F0514-4011 with the MD-3EFJ MET conformation. According to the PLIP results, a residue was marked “+” when at least one protein-ligand interaction was found, whatever its quality (hydrophobic, H-bond, π -stacking, ionic, etc.) and marked by “-” when no protein-ligand interaction was found.
Table 8. List of the protein residues interacting with a nM. inhibitor from the PDB complexes of Table 7 ranked by their number of occurrence. In bold, the residues also found in the interactions with F0514-4011 with the MD-3EFJ MET conformation. According to the PLIP results, a residue was marked “+” when at least one protein-ligand interaction was found, whatever its quality (hydrophobic, H-bond, π -stacking, ionic, etc.) and marked by “-” when no protein-ligand interaction was found.
PDB IDs
Residue4EEV2WGJ5EYD3ZXZ2RFS3RHK3QTI3Q6W2WKM3DKF3LQ8
MET 1160 +++++++++++
LEU 1157 +++-+++++++
ASP 1222 +-+++-+--++
ALA 1108 +-++--++-++
TYR 1230 -++++-+-++-
VAL 1092 -+-+-+++-+-
ILE 1084 ++++ +++-++
TYR 1159 -------++-+
PRO 1158 -+--++--+--
LEU 1140 ----+--++-+
ALA 1221 ------++-+-
PHE 1223 +----+----+
ASP 1164 --------++-
LYS 1110 +----+-----
ASN 1209 --------+--
GLU 1127 ----------+
PHE 1134 +---------+
VAL 1139 +---------+
PHE 1200 +---------+
ARG 1086 -------+---
ARG 1208 -------+---
THR 1343 ------+----
GLU 1347 ------+----
PHE 1089 -----+-----
ASP 1231 ---+-------
ARG 1166 ---+-------
ASN 1167 ---+-------
ILE 1130 +----------
ASN 1171 -----------

Share and Cite

MDPI and ACS Style

Bresso, E.; Furlan, A.; Noel, P.; Leroux, V.; Maina, F.; Dono, R.; Maigret, B. Large-Scale Virtual Screening Against the MET Kinase Domain Identifies a New Putative Inhibitor Type. Molecules 2020, 25, 938. https://doi.org/10.3390/molecules25040938

AMA Style

Bresso E, Furlan A, Noel P, Leroux V, Maina F, Dono R, Maigret B. Large-Scale Virtual Screening Against the MET Kinase Domain Identifies a New Putative Inhibitor Type. Molecules. 2020; 25(4):938. https://doi.org/10.3390/molecules25040938

Chicago/Turabian Style

Bresso, Emmanuel, Alessandro Furlan, Philippe Noel, Vincent Leroux, Flavio Maina, Rosanna Dono, and Bernard Maigret. 2020. "Large-Scale Virtual Screening Against the MET Kinase Domain Identifies a New Putative Inhibitor Type" Molecules 25, no. 4: 938. https://doi.org/10.3390/molecules25040938

Article Metrics

Back to TopTop