Vidushi Sharmaa,
Hirdesh Kumarb and
Sharad Wakode*a
aDepartment of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), University of Delhi, Mehrauli-Badarpur Road, Pushp Vihar, Sector-3, New Delhi – 110017, India. E-mail: sharadwakode@gmail.com; Fax: +91-011-29554503; Tel: +91-9891008594
bParasitology – Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
First published on 4th August 2016
Phosphodiesterase 4B (PDE4B) hydrolyses cyclic adenosine monophosphate (cAMP) and thus regulates its intracellular levels. The enzyme has been proposed as a potential drug target against diseases like inflammation and chronic obstructive pulmonary disease. But use of current PDE4B inhibitors is limited due to dose-dependent nausea and vomiting. Adverse effects associated with current PDE4B inhibitors are possibly results of PDE4D inhibition, a highly similar homolog of PDE4B. Here we considered quinoline analogs and applied ligand-based pharmacophore and atom based 3D-QSAR modeling with structure-based docking and ADME approach. A 5-point pharmacophore model was developed and used to derive a predictive 3D-QSAR model for the studied dataset. The obtained r2 and q2 values were 0.96 and 0.91, respectively. The result suggested that the generated 3D-QSAR model is reliable and can be considered for PDE4B activity prediction. Further, a pharmacophore model was employed for virtual screening to identify potent PDE4B inhibitors. The selective ligands for PDE4B were identified through docking and prime binding energy analysis of ligands in both PDE4B and PDE4D. ADME analysis was performed to confirm the drug ability of the selective ligand. To validate docking results, a molecular dynamics simulation was performed for PDE4B complexed with a top scoring ligand, AQ-390/42425549. AQ-390/42425549-PDE4B interactions reported in MD analysis were consistent with the docking results. All the hit molecules were procured and biologically evaluated for percentage inhibition of PDE4B and PDE4D in in vitro enzymatic assays. Among the total of thirteen molecules that were active against PDE4B, ten were selective with little PDE4D inhibition.
The PDE4 enzyme family consists of 4 members (PDE4A, PDE4B, PDE4C and PDE4D) that are majorly expressed in neutrophils, monocytes, central nervous system (CNS) and smooth muscles of the lung.12–15 Rolipram, non-selective PDE4 inhibitor is reported with severe side effects like nausea and emesis. Second generation PDE4 inhibitors like roflumilast came out with lesser side effects but could not overcome narrow therapeutic window.16,17 Manning et al. proposed that the side effects of PDE4 inhibitors is due to the non-selective inhibition of PDE4 isoforms18 and hence selective inhibition of PDE4 subtypes would be beneficial to improve therapeutic effect to side effect ratio. Previously, several research groups have studied that deletion of PDE4B gene resulted in significant downfall of lipopolysaccharide (LPS)-induced TNF-α production in circulating monocytes and peritoneal macrophages.19,20 Further, the behavioral correlation of emesis in mice by deleting PDE4D encoding gene concluded that inhibition of PDE4D and not PDE4B is responsible for emetic effect of non-selective PDE inhibitors.21 Therefore, selective PDE4B inhibitors can provide anti-inflammatory efficacy without side-effect. However, conserved active site residues of PDE4B and PDE4D make it difficult to design selective PDE4B inhibitors.
In the line of identifying selective PDE4B inhibitors, using the computational techniques, Guariento et al. attempted a ligand-based comparative molecular fields analysis (CoMFA) study for selective PDE4B inhibitors.22 Tripuraneni et al. worked on pharmacophore and atom based 3D-QSAR studies of pyrozolo[1,5-a]pyridine/4,4-dimethylpyrazolone analogues followed by molecular docking and molecular dynamics to design novel molecules with better PDE4 inhibitory activity.23 Dong et al. applied Hopfinger's receptor-dependent QSAR (RD-QSAR) formalism and the Lukacova–Balaz scheme and initiated studies on structure-based multimode QSAR (SBMM QSAR) method, where the structure of the target protein is used in characterizing the ligand, and the multimode issue of ligand binding is systematically treated with a modified Lukacova–Balaz scheme.24 Yang et al. conducted CoMFA studies on 315,6-dihydro-(9H)-pyrazolo-[4,3-c]-1,2,4-triazolo-[4,3-α]-pyridine analogs with variable inhibition of PDE4 to develop models for establishing three-dimensional quantitative structure–activity relationships (3D-QSAR). Comparative molecular field analysis (CoMFA) was conducted on the group of 5,6-dihydro-(9H)-pyrazolo-[4,3-c]-1,2,4-triazolo-[4,3-α]-pyridine analogs to determine the structural requirements for potency in inhibiting PDE4.25 Srivani et al. developed 3D-QSAR with 44 derivatives of triarylethanes to drive structural requirement for PDE4 enzyme inhibition.26 Chakraborti et al. analysed a series of 29 thieno[3,2-d]pyrimidines with PDE4 activity and subjected 3D-QSAR studies using CoMFA and CoMSIA.27 Paola Fossa et al. produced rational pharmacophoric model of the PDE4 enzyme active site using DISCO approach with structurally diverse compounds.28
Here, pharmacophore and 3D-QSAR modeling techniques were used to identify potent and selective PDE4B inhibitors.29,30
The basis of such techniques is that the compounds interacting with the same target could share similar structural or physicochemical properties. A series of quinolines reported as selective PDE4B inhibitors, were used in the present study for developing pharmacophore and 3D-QSAR model.31,32 The quinoline framework was considered as it has emerged as template for the design and identification of novel anti-inflammatory agents.33 The developed models gave useful information of lead optimization for future rational design of PDE4B inhibitors and will be helpful in development of selective inhibitors. The best quantitative model was used as a 3D search query for screening the Specs database to identify new inhibitors of PDE4B. Once identified, the candidate compounds were subsequently subjected to filtration by molecular docking. The selectivity of filtered ligands was optimised by docking in both PDE4B and PDE4D (Fig. 1). ADME analyses were performed to confirm the drug ability of selective ligand.
The resulting pharmacophores were scored and ranked. The scoring was done to identify the best hypothesis, which provided an overall ranking of all the hypotheses. The scoring algorithm included the contributions from the alignment of site points and vectors, volume overlap, selectivity, number of ligands matched, relative conformational energy, and activity. After the careful analyses of the scores and alignment of the active ligands to the generated hypothesis, a best pharmacophore hypothesis was selected.
For validation of the developed hypothesis, a common dataset of 27 active35–38 and 260 inactive molecules was used. Inactive molecules were chosen such that they consists of similar physiochemical descriptors (molecular weight, number of rotational bonds, hydrogen bond donor count, hydrogen bond acceptor count and octanol–water partition coefficient) to active molecules but deprived of any of the chemical descriptors of the active ligands.
Various statistical parameters such as accuracy, precision, sensitivity, specificity and enrichment factor (E value) were calculated for each hypothesis.
Furthermore, for the analysis of results, E value score was calculated using the following formula
For the purpose of QSAR development, van der Waals' models of the aligned training set molecules were placed in a regular grid of cubes, with each cube allotted zero or more ‘bits’ to account for different types of atoms in the training set that occupy the cube. This representation gives rise to binary-valued occupation patterns that can be used as independent variables to create partial least-squares (PLS) QSAR models. QSAR models were generated for the selected hypothesis using the 40-member training set using a grid spacing of 1.0 Å. The best QSAR model was validated by predicting activities of the 8 test set compounds and 1 outlier. A four component (PLS factor) model with good statistics was obtained for the dataset whereas the maximum number of PLS factors in each model can be 1/5 of the total number of training set molecules. Further increase in the number of PLS factors did not improve the model statistics or predictive ability.
For further validation of QSAR model, external r2 prediction was carried out with the data set of thirty two reported PDE4B inhibitors of diverse scaffolds35–39 having activity ranging from 5.0 to 10 pIC50. The reported IC50 values were converted to pIC50 values. The dataset was prepared in Ligprep and then imported to search for matches panel of Phase keeping atom based QSAR model as query. The obtained hits were analysed for their predicted pIC50 values.
Low-energy conformations of ligands that were used for docking program Glide were generated via Ligprep 3.0 with Epik 2.8 of Schrodinger. The optimised structures were produced based on force field OPLS_2005, with protonation states generated at target pH 7.0 ± 2.0. Thirty-two stereoisomers computed by retaining specified chirality and keeping low energy ring conformation 1 per ligand.
Docking was carried out to increase the reliability of the pharmacophore-based screening and to discriminate between the active–inactive and selective ligands for PDE4B. Prepared small molecules were docked into the protein structure using Glide 6.3 XP.42 The score function of Glide or Glide score is a modified and expanded version of ChemScore, was used for binding affinity prediction and ligand ranking. The docking can be on the level of either standard (SP) or extra precision (XP). The addition of large desolvation penalties to both ligand and protein, assignment of specific structural motifs that contribute significantly to binding affinity, and expanded sampling algorithm makes XP superior over SP. In this study, extra precision docking was applied, and the rest of the parameters like ‘dock flexibly’, ‘add epik state penalties to the docking score’ were kept default. The scaling factor was 0.8, and the partial charge cutoff was 0.15. The 3D complex structures of all hits docked in PDE4B and PDE4D were analysed for Glide score and H-bonding interactions. The molecules with considerable difference in docking score and H-bonding interaction in PDE4B and PDE4D were selected and evaluated for binding free energy calculation of docked complex using Prime-MM-GBSA-3.6, employing VSGB continuum dielectric model as solvent model.
The fluorescent polarization (FP) was measured in the Microtiter plate (Black, low binding NUNC Microtiter plate) using a fluorescence reader (TECAN Infinite F200 Pro Microplate Reader) set for excitation at wavelengths ranging from 475–495 nm and detection of emitted light ranging from 518–538 nm. A blank value was subtracted from all other values. The FP value for substrate control wells set as 0% activity while positive control wells set as 100% activity. The % activity for test compound wells were calculated with respect to the positive and substrate control wells. % inhibition was determined by subtracting individual % activity values from 100.
Likewise, PDE4D inhibition was separately calculated in similar experimental setup using 20 μL of PDE4D2 (2.5 pg μL−1) subtype enzyme instead.
![]() | ||
Fig. 2 (A) Pharmacophore model generated for PDE4B. Brown rings (R9 and R10): aromatic rings. Blue sphere (D7): hydrogen-bond donor and pink sphere (A3 and A5): hydrogen-bond acceptor. (B) Common pharmacophoric sites of active ligand with distance. All distances are in Å unit. (C) Alignment of all ligands (quinoline analogues) to the bioactive conformation of crystal ligand 066 of PDB ID 3GWT. (D) Alignment of all active ligands to the pharmacophore. (E). Alignment of all inactive ligands to the pharmacophore. Atom-based PDE4B 3D-QSAR models visualized for (F) positive effect (blue cubes) of electron withdrawing group/atom position (W+) mapped on the most active ligand, (G) negative effect (red cubes) electron withdrawing group/atom mapped position (W−) on the least active ligand, (H) negative effect (red cubes) and positive effect (blue cubes) of position hydrophobic groups (H+,−), (I) positive effect (blue cubes) of position H-bond donor (D+), (J) negative effect (red cubes) of position H-bond donor (D−), (K) negative effect (red cubes) of position of positive ionic groups (P−), (L) negative effect (red cubes) of position of negative ionic groups (N−). |
A four-PLS factor model with good statistics and predictive ability was generated for the dataset (ESI Table 2†). The number of PLS factor included in model development is four as incremental increase in the statistical significance and predictivity was observed for each incremental increase in the incorporated PLS factors up to four. The model expressed r2 value 0.95 exhibited by quinoline based derivatives, is close to one and signifying an acceptable agreement of fitting points on the regression line for the observed and PHASE-predicted activity that is shown in Fig. 3A and B and is summarized in Table 2.
![]() | ||
Fig. 3 (A) Observed and predicted activities of test set compounds associated with PDE4B. (B) Observed and predicted activities of training set compounds associated with PDE4B. |
ID | # factors | SD | r2 | F | Stability | RMSE | q2 | Pearson-R | P |
---|---|---|---|---|---|---|---|---|---|
a SD-standard deviation of the regression; r2-squared value of r for the regression; F-variance ratio (large values of F indicate a more statistically significant regression); P-significance level of variance ratio (smaller values indicate a greater degree of confidence); RMSE-root-mean-square error; q2-squared value of q for the predicted activities; Pearson-R-value for the correlation between the predicted and observed activity for the test set. | |||||||||
AADRR.1108 | 1 | 1.29 | 0.47 | 33 | 0.57 | 0.57 | 0.81 | 0.93 | 1.38 × 10−6 |
2 | 0.86 | 0.77 | 60.21 | 0.67 | 0.46 | 0.88 | 0.95 | 3.29 × 10−12 | |
3 | 0.54 | 0.91 | 120.43 | 0.81 | 0.52 | 0.84 | 0.98 | 1.68 × 10−18 | |
4 | 0.39 | 0.95 | 185.31 | 0.93 | 0.39 | 0.91 | 0.99 | 1.42 × 10−22 |
Generated hypotheses were assessed using dataset of active (27) and inactive (260) ligands. Dataset from literature was employed to validate the generated pharmacophore hypotheses. To each models, different statistical parameters like accuracy, precision, sensitivity, and specificity of the best pharmacophore models were calculated. Furthermore, an E value of 1 was calculated for model (Table 3). From the overall validation results, we assured that hypotheses can differentiate between the actives and inactive molecules.
S. No. | Parameters | Hypothesis AADRR.1108 |
---|---|---|
1 | Total compounds in database (D) | 287 |
2 | Total number of actives in database (A) | 27 |
3 | Total hits (Ht) | 210 |
4 | Active hits (TP) | 22 |
5 | True negative (TN) | 215 |
6 | Enrichment factor or enhancement (E) | 1.1 |
7 | False negatives (FN = A − TP) | 5.0 |
8 | False positives (FP = Ht − TP) | 188 |
9 | Accuracy = (TP + TN)/(TP + TN + FP + FN) | 0.6 |
10 | Precision = TP/(TP + FP) | 0.1 |
11 | Sensitivity = TP/(TP + FN) | 0.8 |
12 | Specificity = TN/(TN + FP) | 0.5 |
The external r2 prediction with the reported PDE4B inhibitors was carried out to validate the QSAR model. The data set of thirty two molecules was taken as an input for searching matches against the developed model in Phase panel. The output window showed twenty molecules as hits. The molecules with the reported and predicted pIC50 values are given in ESI Table 3.† The predicted activity and experimental activity are in statistical limits of confidence and hence the derived model can be accepted for further studies.
The atoms visualize 3D characteristics of the ligands (atoms or pharmacophores) as that contribute positively or negatively to activity. The QSAR model displays 3D characteristics as cubes that represent the model and color according to the sign of their coefficient values, which is indicated as blue for positive coefficients and red for negative coefficients. Positive coefficients indicate an increase and negative coefficients as decrease in activity. This might provide the information that which functional groups/atoms are desirable or undesirable at certain positions in a molecule. The blue cubes in 3D plots of the 3D pharmacophore regions indicates the ligand regions in which the specific feature is important for better activity, whereas the red cubes shows that particular structural feature or functional group that is not essential for the activity or likely the reason for decreased binding potency or affinity.
Visual analysis of Fig. 2F demonstrates that the presence of the blue cubes at the A3, A5, –NH linker and –NHCO group of dimethyl carboxamide and carboxamide substituent, methoxy group and sulphone group pointing out the positive potential of electron withdrawing characteristic of the molecules and is requisite for the activity at this particular place.
It is also apparent from the literature data that the replacement of electron withdrawing groups at the A3-sulphone group (W_3, W_4, W_1) to sulphide (W_6) or amide (W_7, W_8) or removal (W_5) lead to decline in activity. The phenyl sulphones are observed to be better fit to solvent filled pocket of PDE4B active site.
If 3-methoxyphenyl (W_3) was replaced with 3-ethylphenyl (W_11) or cyclohexyl (W_14) or phenyl (W_13), the activity was decreased as methoxy phenyl group was reported to be good fit to the surface.
3-Dimethylcarboxamide group (W_20) attached to phenyl ring, if replaced with 4-methoxy group (W_18) or H (W_3) or 4-t-butyl (W_19) or 4(3-furyl, W_21), all replacements resulted in leap in potency. This was observed that the meta-CONMe2 develops significant van der Waals' contacts with Ser454 and desolvation effect by displacing trapped water molecules.
Likewise replacement of –NH linker (L_15) with –O atom (L_4), –S atom (L_5) and –NMe (L_23) lead to decrease in potency. The role of this –NH linker is reported in developing intramolecular H-bonding with 3-carboxamide substituent at quinoline ring.
Apart from this, 3-carboxamide substituent to quinoline ring was observed to be involved in H-bonding with Asp567, Gln615 and water molecules. Hence its replacement with –CN (L_6), –CH2NH2 (L_7), –COOH (L_8), –CONHMe (L_9), –H (L_12) or with oxadiazoles (L_10 and L_11) produced detrimental effect on the biological activity.
The negative impact of electron withdrawing groups was mapped on inactive ligand to produce remarkable picture. The red cubes in vicinity of –N atom of quinoline ring, –NH linker and sulphone group indicates that presence of electronegative atom in close to these group may produce leap in activity Fig. 2G.
Fig. 2H demonstrates the blue cubes and hence positive impact of hydrophobic groups on PDE4B inhibition and the results are supported by the evident potencies. It can be deduced from the figure and reported as well that hydrophobicity of quinoline ring (pi–pi cloud with Phe618), methyl substituent at quinoline ring (van der Waals contact with Met603), methoxy benzene ring (interaction with protein surface) and benzene ring attached to sulphone ring (interaction with solvent filled pocket) produced increase in biological activity. The substitution of hydrophobic groups near –NH linker to methoxy benzene is unacceptable (L_23) as shown by red cubes or may hinder the binding of the molecules to the receptor active site and will result in decreased PDE4B inhibition.
Fig. 2I illustrates that H-bond donor characteristics is necessary at D7 and –NH linker as consistent with the trend of activity of L_4-12. The red cubes (Fig. 2J) in proximity to D7, –NH linker (L_23) and A5 (L_9) demonstrate negative potential of extension of H-bond on those positions.
Further, Fig. 2K showed negative effect of positive ionic group in replacement with or near to methoxy substituent (red cubes) supported by the reported potencies of W_11, W_14-17. Also Fig. 2L shows the negative impact of negative ionic group near –N atom of quinoline ring and A5 (red cubes) consistent with the decreased biological activity of L_8 and L_6.
The active site residues are mostly conserved between PDE4B and PDE4D. Beside catalytic Gln615 (Gln535 in PDE4D), other H-bonding residues in PDE4B active site like Tyr405 (Tyr325 in PDE4D), His406 (His326 in PDE4D), His410 (His330 in PDE4D), Asp447 (Asp367 in PDE4D), Asp564 (Asp484 in PDE4D), Asn567 (Asn487 in PDE4D) are also conserved between two closely related homologs. Likewise, the hydrophobic residues are also conserved.
Besides structurally resolved catalytic domain, full PDE4 proteins (all PDE4A-4D) also consist of upstream regulatory regions (UCR1-2). Recently, Burgin et al. showed that UCR2 closing over to PDE4B active site region prevent access to substrate ATP.41 Tyr274 of PDE4B-UCR2 is occupied by Phe196 of PDE4D-UCR2 which could make H-bond with the bound inhibitor and therefore provide selective PDE4B inhibition, which can be explored to design selective PDE4B inhibitors. Therefore, in our study we used structures with these additional resolved residues (PDE4B:PDE4D::3G45:3G4G) to perform docking and molecular dynamics study.
Most of the current PDE4B inhibitors are associated with PDE4D related adverse-effects. To minimise PDE4D related toxicity, molecules obtained with glide score < −8 were next docked in PDE4D structure (with resolved N-terminus residue, PDB ID 3G4G) and molecules with similar (or high) docking score in PDE4D structures were selected for further analyses. Molecules with minimum glide score difference of ≥1.5 (PDE4D < PDE4B) were only chosen as selective PDE4B inhibitors.
The ligands exploited the catalytic cavity of PDE4B to much extent and showed H-bond interaction with key residues e.g. Gln615, Tyr274, Asp564, Asn277, His406, Met519 and Ser614. Besides the H-bond, hydrophobic interaction (pi–pi) was established with Tyr405, Tyr274, His450, His406, F586 and F618. In addition to this, metal interaction with Mg2+ was also observed in some of the ligands. Whereas the intrinsic ligands cAMP, and reported inhibitors such as rolipram, roflumilast and 3-isobutyl-1-methylxanthine (IBMX) showed similar glide score, H-bond and hydrophobic ambience in both PDE4B and PDE4D, the screened ligands showed better selectivity profile for PDEB in terms of glide score, H-bond and pi–pi interaction. The ligands could develop H-bond in only three ligands with Tyr325 and Asn375. Phe538 interacted with ionic bond while metal interaction was observed with Mg2+. Pi–pi hydrophobic interaction was associated with Phe538, Phe506, His326 and Tyr325. Above all, while most of the ligands is found to possess either H-bond or pi–pi interaction with Tyr274 of N-terminal of PDE4B, none of the ligands interacted with N-terminal of PDE4D i.e. no interaction with Phe196 of PDE4D, showing selective profile of screened ligands.
54 ligands were finalized based on considerable difference in docking score of both PDE4B and PDE4D. 27 ligands were selected for considerable difference in Prime binding energy of PDE4B and PDE4D docked complexes.
The ADME properties of all the obtained ligands were assessed using Qikprop 4.0, and these ADME properties of best hits are listed in ESI Table 4.† The 21 of the hits showed optimum ADME properties while 6 were rejected. The ADME property of these hits makes them promising candidates for as PDE4B inhibitors. The selective inhibitors, their glide scores along with the hydrogen bonding of key residues are presented in Table 4 and ESI Table 5.† The binding pose of top scoring ligand is depicted in Fig. 4A.
Molecule | G score | H-Bond residues | ΔG binding of complex | |||
---|---|---|---|---|---|---|
PDE4B | PDE4D | PDE4B | PDE4D | PDE4B | PDE4D | |
AQ-390/42425549 | −11.8 | −8.7 | Gln615, Tyr274 | No | −96.5 | −77.6 |
AG-690/11972161 | −11.7 | −8.7 | Gln615 | No | −95.5 | −82.3 |
AO-476/43407280 | −11.3 | −9.3 | Gln615 | No | −88.6 | −74.1 |
AG-690/36276051 | −11.3 | −7.8 | Gln615 | No | −76.4 | −65.7 |
AO-022/43390834 | −11.3 | −4.7 | Tyr274, Asp564 | No | −27.6 | −10.4 |
AG-205/36564043 | −11.3 | −8.5 | His406 | No | −30.5 | −4.1 |
AG-690/36276042 | −11.1 | −8.2 | Gln615 | No | −66.8 | −46.6 |
AG-690/36873050 | −10.9 | −8.8 | Tyr405, Asn455 | No | −57.3 | −30.9 |
AJ-292/42152568 | −10.6 | −7 | Asp564 | Asn375 | −72.5 | −63.6 |
AF-399/42316263 | −10.5 | −5.7 | Gln615 | No | −64.9 | −51.0 |
AK-968/37005156 | −10.4 | −8.4 | Gln615 | No | −83.9 | −74.9 |
AO-022/43453692 | −10.3 | −8.1 | Tyr405, Tyr274 | Tyr325 | −92.2 | −78.3 |
AF-399/41980308 | −10.2 | −8 | Gln615 | No | −55.9 | −43.0 |
AP-964/40915318 | −10.0 | −8.1 | Tyr274, Met519, Asn277 | No | −55.9 | −18.7 |
AG-205/33161053 | −10.1 | −7.6 | Gln615, Tyr274, Ser614 | No | −34.8 | −21.7 |
AT-057/43469096 | −10 | −5.3 | Tyr274 | No | −76.4 | −39.1 |
AG-690/15437723 | −9.7 | −5.3 | Gln615, Tyr274, His450 | No | −59.2 | −43.9 |
AK-968/12386394 | −9.7 | −5.9 | Gln615 | No | −69.5 | −57.8 |
AF-399/15335138 | −9.6 | −7.4 | Tyr274 | No | −64.3 | −50.8 |
AG-690/10252051 | −9.4 | −7.7 | Gln615 | No | −96.6 | −85.5 |
AP-124/43383688 | −9.2 | −7.5 | Tyr274 | No | −57.7 | −41.3 |
Rolipram | −9.2 | −9.3 | Gln615 | No | −81.2 | −83.4 |
IBMX | −7.3 | −7.7 | Gln615 | Gln535 | −51.5 | −46.8 |
Roflumilast | −9.7 | −9.6 | Gln615 | Gln535 | −71.1 | −79.1 |
cAMP | −8.2 | −8.6 | Gln615 | Asp438, Asn199, Asn375 | −43.7 | −44.4 |
![]() | ||
Fig. 4 (A) Docked pose of AQ-390/42425549 (AQ) in PDE4B (PDB ID 3G45). The molecule showed H-bond with Tyr274 and Gln615 (blue solid line) and pi–pi interaction with Phe618. These interactions were also observed in the molecular dynamics simulation of PDE4B-AQ complex. The values represents the average and standard deviation of different distances during last 3 ns of MD simulation. (B) RMSD plot of protein and ligand for the 10 ns MD simulation run. |
Molecules | % inhibition of PDE4B | % inhibition of PDE4D |
---|---|---|
a n.d.: not determined. | ||
AQ-390/42425549 | 73.16 | 30.39 |
AG-690/11972161 | 65.98 | 26.96 |
AG-690/36276051 | 63.93 | 39.95 |
AG-205/36564043 | 68.24 | 37.75 |
AG-690/36276042 | 70.49 | 36.52 |
AP-964/40915318 | 61.27 | 30.39 |
AG-205/33161053 | 71.72 | 38.73 |
AG-690/15437723 | 77.87 | 39.46 |
AK-968/12386394 | 65.37 | 39.71 |
AF-399/15335138 | 70.08 | 37.01 |
AJ-292/42152568 | 78.48 | 59.56 |
AO-022/43453692 | 60.25 | 56.13 |
AP-124/43383688 | 63.93 | 67.16 |
AG-690/36873050 | 30.94 | n.d. |
AF-399/42316263 | 29.10 | n.d. |
AK-968/37005156 | 31.15 | n.d. |
AF-399/41980308 | 40.57 | n.d. |
AT-057/43469096 | 46.72 | n.d. |
AG-690/10252051 | 39.34 | n.d. |
AO-476/43407280 | 51.43 | n.d. |
AO-022/43390834 | 44.88 | n.d. |
Finally, 21 potential molecules were identified with PDE4B selectivity using virtual screening, docking and ADME properties analysis. Molecular dynamics study was carried out for a top scoring ligand, AQ and interaction pattern of ligand protein complex was found to be consistent with the docking results. All the hits were purchased and biologically evaluated for percentage inhibition of PDE4B and PDE4D with in vitro enzymatic assay. Out of these, thirteen hits have shown potent and ten showed selective inhibitory activity against PDE4B. All this data provides confidence level in the present study to find its application for designing the potent and selective PDE4B inhibitors.
Footnote |
† Electronic supplementary information (ESI) available: List of all hypotheses produced. List of all quinoline analogues with their QSAR-model set and pharma set classification. Predicted absorption, distribution, metabolism and excretion (ADME) properties of all PDE4B selective molecules. Chemical structure of all PDE4B selective molecules and their Specs IDs. Data for molecular dynamics simulation and table for external r2 prediction of QSAR model. See DOI: 10.1039/c6ra11210b |
This journal is © The Royal Society of Chemistry 2016 |