Next Article in Journal
Four Chemotherapeutic Compounds That Limit Blood-Brain-Barrier Invasion by Toxoplasma gondii
Previous Article in Journal
Synthesis and Antifungal Activity of Chimonanthus praecox Derivatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations

by
Amany Belal
1,*,
Hazem Elkady
2,
Ahmed A. Al-Karmalawy
3,
Ali H. Amin
4,5,
Mohammed M. Ghoneim
6,7,
Mohamed El-Sherbiny
8,
Rasha Hamed Al-Serwi
9,
Mohamed Attia Abdou
10,
Mona H. Ibrahim
11 and
Ahmed B. M. Mehany
12,*
1
Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
2
Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
3
Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
4
Deanship of Scientific Research, Umm Al-Qura University, Makkah 21955, Saudi Arabia
5
Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
6
Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
7
Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Riyadh 13713, Saudi Arabia
8
Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
9
Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
10
Orthopedic Surgery Department, Al Iman General Hospital, Riyadh 11671, Saudi Arabia
11
Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11884, Egypt
12
Zoology Department, Faculty of Science (Boys), Al-Azhar University, Cairo 11884, Egypt
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(17), 5571; https://doi.org/10.3390/molecules27175571
Submission received: 15 July 2022 / Revised: 13 August 2022 / Accepted: 18 August 2022 / Published: 30 August 2022

Abstract

:
Bone morphogenetic proteins (BMPs) are growth factors that have a vital role in the production of bone, cartilage, ligaments, and tendons. Tumors’ upregulation of bone morphogenetic proteins (BMPs) and their receptors are key features of cancer progression. Regulation of the BMP kinase system is a new promising strategy for the development of anti-cancer drugs. In this work, based on a careful literature study, a library of benzothiophene and benzofuran derivatives was subjected to different computational techniques to study the effect of chemical structure changes on the ability of these two scaffolds to target BMP-2 inducible kinase, and to reach promising candidates with proposed activity against BMP-2 inducible kinase. The results of screening against Lipinski’s and Veber’s Rules produced twenty-one outside eighty-four compounds having drug-like molecular nature. Computational ADMET studies favored ten compounds (11, 26, 27, 29, 30, 31, 34, 35, 65, and 72) with good pharmacokinetic profile. Computational toxicity studies excluded compound 34 to elect nine compounds for molecular docking studies which displayed eight compounds (26, 27, 29, 30, 31, 35, 65, and 72) as promising BMP-2 inducible kinase inhibitors. The nine fascinating compounds will be subjected to extensive screening against serine/threonine kinases to explore their potential against these critical proteins. These promising candidates based on benzothiophene and benzofuran scaffolds deserve further clinical investigation as BMP-2 kinase inhibitors for the treatment of cancer.

Graphical Abstract

1. Introduction

Bone morphogenetic proteins (BMPs) are a type of transforming growth factor-β (TGF-β) superfamily that has multiple functions. More than a quarter of a million BMP members have been identified. Cell proliferation, survival, differentiation, and apoptosis are all regulated by BMPs. It is important to produce bone, cartilage, ligaments, and tendons [1].
When BMPs attach to their cell surface receptors on mesenchymal cells, the BMP signaling cascade is initiated, and signals are transmitted to the cell nucleus via particular proteins. The mesenchymal cell becomes a chondrocyte or an osteoblast as a result of the expression of genes that lead to the production of macromolecules involved in cartilage and bone development [2]. Following the implantation of this protein component of the bone matrix, a complex series of cellular events occurred, including mesenchymal cell infiltration, cartilage formation, vascularization, bone formation, and finally remodeling of new bone tissue, as well as population by hematopoietic bone marrow elements [3]. BMPs have been found to directly differentiate cells into the osteoblast phenotype, in addition to chondrocyte lineage differentiation [4]. Bone morphogenetic protein-2 (BMP-2) is the only osteoinductive growth factor approved by the FDA for use as a bone graft alternative.
BMP signaling is associated with cancer-related cellular phenotypes. BMPs can increase cell migration and invasiveness by arresting the cell cycle of many different cell types in the early G1 phase [5]. BMPs have been shown to play a function in a wide range of cancers and other malignancies. BMPs can either suppress or promote tumorigenesis, with the majority of cases favoring metastasis [6]. Intestinal tumorigenesis can be triggered by disruption of the tightly controlled homeostatic BMP signaling gradients [7]. The most important factor of hereditary risk and predisposition for sporadic colorectal cancer (CRC) susceptibility, according to recent research, is variation in the BMP pathway [8]. As rhBMP-2 has been observed to be associated with a higher risk of developing new cancer than vertebral bone graft, its use in spine surgery has been the subject of significant discussion [9]. BMP-2-inducible kinase belongs to the Numb-associated kinase (NAK) family of serine/threonine kinases [10]. BMP-2 is crucial to the occurrence and progression of colon cancer, prostatic carcinoma, and lung cancer [11,12]. Furthermore, BMP-2 is essential for prenatal and postnatal mammary gland development, but it has also been identified in breast cancer cells [13]. Additionally, BMP-2 increases gastric cancer cell motility and invasion by activating PI-3 kinase/Akt; blocking this pathway may limit BMP-2-mediated metastasis [14]. Various heterocyclic molecules are found in a variety of drugs and have become a key research foundation in medicinal chemistry. This is mostly owing to the adaptability and specific physicochemical properties of heterocyclic molecules. Benzofuran is one of the identified heterocyclic compounds [15]. Benzothiophene and Benzofuran scaffold is one of the privileged frameworks in drug development, as this core displays diverse biological activities allowing them to function as anti-convulsant, anti-cancer, anti-diabetic, anti-tubercular, anti-oxidant, anti-inflammatory, anti-microbial, and many other agents [16,17].
Many literature studies revealed that substituted benzothiophene and benzofuran derivatives were a new class of small molecules that act as potential anabolic agents targeting BMP-2 [18,19]. In particular, compounds I, II III, and IV (Figure 1) enhanced BMP-2 expression in vitro and stimulated bone formation and trabecular connectivity restoration in vivo [18]. In contrast, benzothiophene and benzofuran derivatives can inhibit several protein kinases and act as anticancer agents such as compounds VVIII (Figure 2) [16,20,21,22].
In this work, we will try to find new inhibitors for BMP-2 that may be useful in the treatment of several types of cancers. We decided to investigate some reported benzothiophene and benzofuran derivatives [23,24] for their potential as BMP-2 inducible kinase inhibitors. The selected library (Figure S1 Supplementary data) was subjected to different computational techniques such as ADMET, toxicity, and docking studies to reach promising candidates with proposed activity against BMP-2-inducible kinase. Finally, the promising compounds will undergo additional screening against kinases related to serine/threonine kinases (CDK2, Pim1, cell division protein kinase 2, casein kinase II, and eukaryotic translation initiation factor 2-alpha kinase 3).

2. Results and Discussion

2.1. Virtual Screening

In the current study, an in silico computational study was conducted to determine the number of rotatable bonds, topology polar surface area (TPSA), and other physicochemical properties for the tested candidates according to the directions of Veber’s and Lipinski’s Rules of five [25].
Lipinski proposed that the absorption of an orally administered compound is more likely to be better if the molecule satisfies at least three out of four of the following rules: (1) hydrogen bond donors (HBD) ≤ 5; (2) hydrogen bond acceptors (HBA) ≤ 10; (3) molecular weight < 500; (4) a coefficient of partition between octanol and water logP < 5. Compounds contravening more than one of these rules could not have good bioavailability. Moreover, reduced molecular flexibility, as measured by the number of rotatable bonds, and low polar surface area are found to be important predictors of good oral bioavailability [26]. In this regard, Veber’s Rule says that a compound with 10 or fewer rotatable bonds (RTB) and a polar surface area (TPSA) no greater than 140 ′Å2 should present good oral bioavailability [26].
The results presented in Table 1 showed that twenty-one outside eighty-four tested compounds (11, 26–35, 40, 61–63, 65, and 68–72) showed no contravention of Lipinski’s and Veber’s Rules, respectively, and hence display a drug-like molecular nature.
At first, with respect to the number of hydrogen bond donors (HBD), compounds 11, 26–35, 40, 61–63, 65, and 68–72 have an HBD range of 0–4. These values are less than 5 and obey the first parameter of Lipinski’s Rule. Moreover, regarding the number of hydrogen bond acceptors (HBA), all the mentioned 21 compounds have HBA ranging from 3–8 (i.e., less than 10), which meets the second parameter Lipinski’s Rule. Concerning the molecular weight parameter of compounds 11, 26–35, 40, 61–63, 65, and 68–72, it was found that these compounds have a molecular weight ranging from 208.234 to 499.604. Such values are less than 500, which confirms the third parameter of Lipinski’s Rule. In addition, compounds 11, 26–34, 65, 68, and 72 demonstrated a logP value range from 1.45 to 4.994. The obtained values match with the fourth parameter of Lipinski’s Rule, which suggests that good drug candidates should have logP values less than 5. According to Veber’s Rule, number of rotatable bonds was calculated, and this is an important parameter to measure the molecular flexibility and oral bioavailability of the drug candidates. The results revealed that compounds 11, 26–35, 40, 61–63, 65, and 68–72 displayed an acceptable number of rotatable bonds ranging from 2–10 that meet the criteria of Veber’s Rule. Moreover, the number of TPSA (a physicochemical property describing the polarity of molecules) of such compounds is within the acceptable values (ranging from 74.77 to 123.91 Å2) of less than 140 Ų. On the other hand, the rest of the tested compounds did not meet the criteria of Lipinski’s and Veber’s Rules, respectively. A closer look at the data presented in Table S1 (Supplementary Data) demonstrated that compounds 1–10, 12–25, 36–39, 41–60, 64, 66, 67, and 73–84 have a molecular weight of more than 500 and logP values of more than 5, which does not meet the criteria of Lipinski’s Rule. Additionally, most of them do not obey Veber’s Rule, as shown in Table S1 (Supplementary Data).

2.2. Computational ADMET Analysis

Twenty-one compounds, 11, 26–35, 40, 61–65, 68–72, met the criteria of Lipinski’s and Veber’s Rules and were further investigated for their pharmacokinetic properties (ADMET studies) according to the reported procedures [27,28,29,30,31,32]. N-[6-(3-[[(cyclopropylmethyl)sulfonyl]amino]phenyl)-1H-indazol-3-yl]cyclopropanecarboxamide (IDK), an indazole inhibitor, is used as a reference molecule in this computational study. Blood–brain barrier (BBB) penetration, aqueous solubility, intestinal absorption, CYP2D6 binding, and plasma protein binding properties of the elected candidates were calculated using Discovery studio 4.0 (Discovery Studio 2016 Vélizy-Villacoublay, France).
The results revealed that ten out of twenty-one compounds showed good ADMET profiles and drug-likeness properties. In detail, compounds 11, 26, 27, 29, 30, 31, 34, 35, 65, and 72 exhibited medium to very low BBB penetration levels (Figure 3); therefore, such compounds were anticipated to be safe for the CNS. In addition, it was found that all of the ten compounds had moderate to good absorption behavior. Moreover, the solubility level of the ten compounds is expected to be better than or even similar to that of the reference drug that showed a low solubility level. For cytochrome P450 2D6 (CYP2D6) inhibition, all the examined members were predicted as non-inhibitors. Finally, all compounds were expected to bind to plasma protein by more than 90% (Table 2).

2.3. Computational Toxicity Studies

Based on the previous findings, computational toxicity profiles of the favored ten compounds (11, 26, 27, 29, 30, 31, 34, 35, 65, and 72) were examined following the reported procedures [27,28,29,30,31,32]. Six toxicity models were used through Discovery studio software version 4.0 [33,34].
All the tested compounds showed TD50 values better than IDK, and the values ranged from 6.036 to 431.948 g/kg body. Furthermore, compounds 27, 29, 30, and 31 showed rat maximum tolerated dose values higher than that of IDK, and the values range is from 0.190 to 0.210 g/kg body weight. Conversely, the rest of the compounds showed maximum tolerated doses ranging from 0.035 to 0.115 g/kg that were lower than that of IDK. Compounds 34, 35, and 65 revealed rat oral LD50 values of 2.364, 5.598, and 5.308, respectively, which were higher than that of IDK (2.626 g/kg body weight). On the other hand, the rest of the compounds showed oral LD50 values ranging from 0.230 to 2.223 g/kg body weight, which were lower than that of IDK. Additionally, all compounds were estimated to be non-toxic against the developmental toxicity potential model except compound 34. Moreover, compounds 26, 27, 34, and 72 showed LOAEL values ranging from 0.053 to 0.088 g/kg body weight whereas IDK exhibited 0.049 g/kg body weight. Finally, all the elected compounds showed no irritancy in the skin irritancy model as shown in Table S2 (Supplementary Data).

2.4. Molecular Docking Studies

Molecular docking was performed using Molecular Operating Environment (MOE) [35] to recognize the binding modes and interactions with the crucial amino acids [36,37,38,39,40,41]. Nine compounds (11, 26, 27, 29, 30, 31, 35, 65, and 72) displayed good computational ADMET and toxicity profiles. They were subjected to further docking studies. Such studies were carried out for the tested candidates to inspect their binding free energies (∆G) and binding modes against BMP-2-inducible kinase (PDB ID: 5I3R) using IDK as a reference (Table 3).
At first, the docking procedure was validated and the RMSD value was 1.05, which indicated the validity of the docking process (Figure S2) (See Supplementary data).
IDK orientation with the amino acids of the pocket has been studied and displayed in Figure 4. The proposed binding pattern revealed an affinity value of −22.72 Kcal/mol. The interaction between IDK and BMP-2-inducible kinase is stabilized through five hydrogen bonds and a series of hydrophobic interactions. The N-(1H-indazol-3-yl)cyclopropanecarboxamide moiety forms three hydrogen bonds with the two crucial amino acids, Cys133 and Glu131, and nine hydrophobic interactions with Cys133, Leu57, Leu187, Val65, and Ala77. Moreover, 1-cyclopropyl-N-phenylmethanesulfonamide moiety was buried in the active pocket of BMP-2-inducible kinase through the formation of two hydrogen bonds with Gln137 and Asn185 and four hydrophobic interactions with Val65, Ala58, and Lys79 (Figure 4).
Compound 26 (affinity value of −22.90 Kcal/mol) combined with the receptor protein in a manner similar to that of IDK. The 3-methoxybenzo[b]thiophene moiety formed two hydrogen bonds with Cys197 and Gln137, in addition to seven hydrophobic interactions with Val65, Cys197, Ala58, Ala77, and Leu187, and two pi-sulfur interactions with Met130. Furthermore, the 2-carboxylic acid moiety formed two hydrogen bonds with the two essential amino acids, Cys133 and Glu131 (Figure 5).
Docking simulation of compound 27 revealed that it has a good fitting into the enzyme active site with a docking score of −23.34 Kcal/mol. The benzo[b]thiophene-2-carboxylic acid moiety formed two hydrogen bonds with Cys133 and Cys197. In addition, it formed seven pi-alkyl and pi-sigma interactions with Ala77, Val65, Cys197, Ala58, and Leu187. Furthermore, the 2-amino-2-oxoethoxy moiety formed two hydrogen bonds with Gln137 (Figure 6).
Interestingly, the docking result of compound 31 (affinity value of −22.22 Kcal/mol) is almost like that of IDK. The 3-ethoxybenzo[b]thiophene-2-carboxylic acid moiety formed three hydrogen bonds with Cys133 and Glu131. In addition, it is involved in binding with the receptor through the formation of seven hydrophobic interactions with Ala77, Ala58, Cys197, Leu187, and Val65, in addition to two pi-sulfur interactions with Met130. On the other hand, the 2-nitrobenzene moiety formed one pi-pi interaction with Leu57 (Figure 7).
Molecular docking results of compound 35 revealed an affinity value of −24.25 Kcal/mol. The results showed formation of five hydrogen-bonding interactions, ten hydrophobic interactions, in addition to one electrostatic interaction, as observed in Figure 8.
The proposed binding mode of compound 72 revealed an affinity value of −24.22 Kcal/mol. The 3-(benzyloxy) thieno[2,3-c]pyridine-2-carboxamide moiety formed ten hydrophobic interactions with Cys133, Ala77, Leu57, Leu187, and Val65. Likewise, it formed two hydrogen bonds with Glu131 and Tyr132, in addition to one pi-sulfur interaction with Met130. Moreover, the N-(3-(thiophene-2-carboxamido)benzyl) moiety formed three extra hydrogen bonds with Glu184, Gly60, and Lys182 and one pi-pi interaction with Ala58 (Figure 9).

2.5. Further Investigation against Protein Kinases

The promising nine compounds will be subjected to further screening against kinases in general to explore their potential against these crucial proteins. Compound 11 was revealed to target 48 types of kinases (Supplementary Data) with a binding affinity ranging from −4.694 to −10.033 Kcal/mol. Compound 11 showed a ligand similarity score of 0.428 with Cell division protein kinase 2 ligand (A27). Moreover, it showed a binding similarity score of more than 66% with a docking score equal to −9.463 Kcal/mol. Figure 10 illustrates the mode and interactions of compound 11 with Cell division protein kinase 2. Compound 26 (Figure 11) revealed good potential to target three types of kinases, Casein kinase II subunit alpha, Proto-oncogene serine/threonine-protein kinase pim-1, and Serine/threonine-protein kinase pim-1 with a ligand similarity score of more than 40% and a binding affinity range from −6.4 to −7.00 Kcal/mol. Compound 27 showed the ability to target seven types of kinases with a ligand similarity score of more than 40% and a binding affinity range from −6.4 to −6.7 Kcal/mol. The best binding affinity was assigned for Serine/threonine-protein kinase pim-1. Furthermore, compound 27 revealed a good potential to target seven kinases with ligand similarity of more than 40% and a binding affinity range from −6.7 to 8.7 Kcal/mol, and it has shown a ligand and binding similarity mode of more than 40% and a docking score energy equal to −8.7 Kcal/mol with Cyclin-dependent kinase 2. Compound 30 showed potential against nine types of protein kinases, and the best proposed potential was assigned for cell division protein kinase 2 with a binding affinity of −8.7 Kcal/mol. Figure 12 exhibits the binding mode and interactions of compound 30 with cell division protein kinase 2.
Compound 31 showed binding affinity equal to −8.53 against Cyclin-dependent kinase 2 and compound 35 showed more binding affinity towards protein kinase named Eukaryotic translation initiation factor 2-alpha kinase, and its binding affinity score is −11.812 Kcal/mol. Figure 13 show 2D binding mode of compound 35 with Eukaryotic translation initiation factor 2-alpha kinase-3. Compounds 65 and 72 revealed no potential to target kinases through the ligTMap tool.

3. Conclusions

Eighty-four benzothiophene and benzofuran derivatives were selected for in silico screening to reach promising BMP-2 inducible kinase inhibitors. Twenty-one compounds showed agreement with both Lipinski’s and Veber’s Rules. In silico ADMET studies revealed ten compounds (11, 26, 27, 29, 30, 31, 34, 35, 65, and 72) with good pharmacokinetic profile. Additionally, in silico toxicity studies revealed nine compounds (11, 26, 27, 29, 30, 31, 35, 65, and 72) with an acceptable toxicity profile. In addition, docking studies were carried out on BMP-2 and other kinases using the nine promising compounds. Molecular docking against BMP-2 inducible kinases demonstrated that eight compounds (26, 27, 29, 30, 31, 35, 65, and 72) with significant binding affinity to the target displayed significant BMP-2 inducible kinase inhibitor properties. Finally, we can say that these organic molecules deserve further pre-clinical and clinical investigations as they may serve as anti-BMP-2 inducible kinases.

4. Experimental Section

4.1. Screening against Lipinski’s and Veber’s Rules

Lipinski and Veber descriptors were determined using Discovery studio 4.0. At first, the CHARMM force field was applied then the tested compounds were prepared and minimized according to the preparation of small molecule protocol. Then, the Lipinski and Veber descriptors protocol was applied to carry out these studies.

4.2. Computational ADMET Studies

ADMET descriptors (absorption, distribution, metabolism, excretion and toxicity) of the compounds were determined using Discovery studio 4.0. At first, the CHARMM force field was applied then the tested compounds were prepared and minimized according to the preparation of small molecule protocol. Then, ADMET descriptors protocol was applied to carry out these studies [42,43,44,45,46,47].

4.3. Computational Toxicity Studies

The toxicity parameters of the synthesized compounds were calculated using Discovery studio 4.0. IDK was used as a reference drug. At first, the CHARMM force field was applied then the compounds were prepared and minimized according to the preparation of small molecule protocol. Then, different parameters were calculated from the toxicity prediction (extensible) protocol [29,48].

4.4. Docking Studies

Crystallographic structure of tubulin [PDB ID: 5I3R, resolution: 2.40 Å] was retrieved from Protein Data Bank (http://www.pdb.org) and considered a target for docking simulation. The docking analysis was performed using MOE2014 software (Montreal, QC, Canada) to evaluate the free energy and binding modes of the synthesized compounds against BMP-2-inducible kinase. At first, the crystal structure of the target was prepared by removing water molecules and retaining the essential chain and the co-crystallized ligand (IDK). Then, the protein structure was protonated, and the hydrogen atoms were hidden. Next, the energy was minimized, and the binding pocket of the protein was defined.
The 2D structures of the tested compounds and reference ligand (IDK) were sketched using ChemBioDraw Ultra 14.0 and saved in MDL-SD format. Then, the saved files were opened using MOE and 3D structures were protonated. Next, energy minimization was applied. Before the docking process, validation of the docking protocol was carried out by running the simulation only using the co-crystallized ligand (IDK), which showed a low RMSD value. The molecular docking of the tested compounds was performed using a default protocol against the target receptor. In each case, 30 docked structures were generated using genetic algorithm searches, ASE was used for scoring, and forcefield (MMFF94X) for refinement. The ASE scoring function estimates the free energy of binding of the ligand from a given pose. The functional form is a sum of terms:
Δ G =   c + E f l e x + h b o n d s c H B   f H B + m l i g c M   f M + a t o m s   i Δ D i
where C represents the average gain/loss of rotational and translational entropy; Eflex is the energy due to the loss of flexibility of the ligand (calculated from ligand topology only); FHB measures geometric imperfections of hydrogen bonds and takes a value in [0,1]; CHB is the energy of an ideal hydrogen bond; FM measures geometric imperfections of metal ligations and takes a value in [0,1]; CM is the energy of an ideal metal ligation, and Di is the desolvation energy of atom i.
The output from MOE was further analyzed and visualized using Discovery Studio 4.0 software [49,50,51,52,53,54,55].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27175571/s1. Figure S1: The chemical structures of the tested compounds; Table S2: Physicochemical properties of the tested compounds not passed Lipinski and Veber Rules; Figure S2: Validation of the docking process showed alignment of the co-crystallized pose and the re-docked pose of the same ligand inside BMP-2-inducible kinase; Supplementary data of toxicity study.

Author Contributions

Conceptualization, A.B.; Data curation, H.E., A.A.A.-K., A.H.A., M.M.G., M.A.A., M.H.I. and A.B.M.M.; Investigation, A.A.A.-K., M.M.G., R.H.A.-S., M.A.A. and A.B.M.M.; Methodology, H.E., A.B. and M.H.I.; Resources, A.H.A. and M.M.G.; Software, H.E. and M.H.I.; Validation, A.A.A.-K., M.E.-S. and M.A.A.; Visualization, A.B., A.H.A., M.E.-S., R.H.A.-S. and A.B.M.M.; Writing—original draft, H.E.; Writing—review and editing, A.B., M.E.-S., R.H.A.-S., A.A.A.-K. and M.H.I. All authors have read and agreed to the published version of the manuscript.

Funding

The Deanship of Scientific Research at Umm Al-Qura University supported this work by Grant Code: (22UQU4331100DSR14). This publication was supported by AlMaarefa University researchers supporting program (grant number: MA-006), AlMaarefa University, Riyadh, Saudi Arabia. The work was supported by Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2022R199), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia. This research work was also supported by Taif University Researchers Supporting Project number (TURSP-2020/35), Taif University, Taif, Saudi Arabia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are supplied in supplementary information files.

Acknowledgments

The authors would like to thank the Deanship of Scientific Research at Umm Al-Qura University for supporting this work by Grant Code: (22UQU4331100DSR14). This publication was supported by AlMaarefa University researchers supporting program (grant number: MA-006), AlMaarefa University, Riyadh, Saudi Arabia. The work was supported by Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2022R199), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia. Amany Belal would like to thank Taif University Researchers Supporting Project number (TURSP-2020/35), Taif University, Taif, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xiao, Y.T.; Xiang, L.X.; Shao, J.Z. Bone morphogenetic protein. Biochem. Biophys. Res. Commun. 2007, 362, 550–553. [Google Scholar] [CrossRef] [PubMed]
  2. Reddi, A.H. Bone Morphogenetic Proteins: From Basic Science to Clinical Applications. J. Bone Jt. Surg. 2001, 83, S1–S6. [Google Scholar] [CrossRef] [PubMed]
  3. Reddi, A. Cell Biology and Biochemistry of Endochondral Bone Development. Collagen Relat. Res. 1981, 1, 209–226. [Google Scholar] [CrossRef]
  4. Jain, A.P.; Pundir, S.; Sharma, A. Bone morphogenetic proteins: The anomalous molecules. J. Indian Soc. Periodontol. 2013, 17, 583–586. [Google Scholar] [CrossRef] [PubMed]
  5. Davis, H.; Raja, E.; Miyazono, K.; Tsubakihara, Y.; Moustakas, A. Mechanisms of action of bone morphogenetic proteins in cancer. Cytokine Growth Factor Rev. 2015, 27, 81–92. [Google Scholar] [CrossRef] [PubMed]
  6. Ehata, S.; Yokoyama, Y.; Takahashi, K.; Miyazono, K. Bi-directional roles of bone morphogenetic proteins in cancer: Another molecular Jekyll and Hyde? Pathol. Int. 2013, 63, 287–296. [Google Scholar] [CrossRef] [PubMed]
  7. Hardwick, J.C.; Kodach, L.; Offerhaus, G.J.; Brink, G.R.V.D. Bone morphogenetic protein signalling in colorectal cancer. Nat. Cancer 2008, 8, 806–812. [Google Scholar] [CrossRef]
  8. Jaeger, E.; Leedham, S.J.; Lewis, A.; Segditsas, S.; Becker, M.; Cuadrado, P.R.; Davis, H.; Kaur, K.; Heinimann, K.; Howarth, K.; et al. Hereditary mixed polyposis syndrome is caused by a 40-kb upstream duplication that leads to increased and ectopic expression of the BMP antagonist GREM1. Nat. Genet. 2012, 44, 699–703. [Google Scholar] [CrossRef]
  9. Skovrlj, B.; Koehler, S.M.; Anderson, P.A.; Qureshi, S.A.; Hecht, A.C.; Iatridis, J.C.; Cho, S.K. Association Between BMP-2 and Carcinogenicity. Spine 2015, 40, 1862–1871. [Google Scholar] [CrossRef]
  10. James, A.W.; Lachaud, G.; Shen, J.; Asatrian, G.; Nguyen, V.; Zhang, X.; Ting, K.; Soo, C. A Review of the Clinical Side Effects of Bone Morphogenetic Protein-2. Tissue Eng. Part B Rev. 2016, 22, 284–297. [Google Scholar] [CrossRef]
  11. Ehata, S.; Miyazono, K. Bone Morphogenetic Protein Signaling in Cancer; Some Topics in the Recent 10 Years. Front. Cell Dev. Biol. 2022, 10, 883523. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, Y.; Ma, L.; Guo, Q.; Zhang, S. Expression of bone morphogenetic protein-2 and its receptors in epithelial ovarian cancer and their influence on the prognosis of ovarian cancer patients. J. Exp. Clin. Cancer Res. 2010, 29, 85–86. [Google Scholar] [CrossRef]
  13. Raida, M.; Clement, J.H.; Ameri, K.; Han, C.; Leek, R.D.; Harris, A.L. Expression of bone morphogenetic protein 2 in breast cancer cells inhibits hypoxic cell death. Int. J. Oncol. 2005, 26, 1465–1470. [Google Scholar] [CrossRef] [PubMed]
  14. Kang, M.H.; Kim, J.S.; Seo, J.E.; Oh, S.C.; Yoo, Y.A. BMP2 accelerates the motility and invasiveness of gastric cancer cells via activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Exp. Cell Res. 2010, 316, 24–37. [Google Scholar] [CrossRef] [PubMed]
  15. Radadiya, A.; Shah, A. Bioactive benzofuran derivatives: An insight on lead developments, radioligands and advances of the last decade. Eur. J. Med. Chem. 2015, 5, 356–376. [Google Scholar] [CrossRef] [PubMed]
  16. Keri, R.S.; Chand, K.; Budagumpi, S.; Somappa, S.B.; Patil, S.A.; Nagaraja, B.M. An overview of benzo [b] thiophene-based medicinal chemistry. Eur. J. Med. Chem. 2017, 29, 1002–1033. [Google Scholar] [CrossRef]
  17. Farhat, J.; Alzyoud, L.; Alwahsh, M.; Al-Omari, B. Structure–Activity Relationship of Benzofuran Derivatives with Potential Anticancer Activity. Cancers 2022, 14, 2196. [Google Scholar] [CrossRef]
  18. Guo, H.-F.; Shao, H.-Y.; Yang, Z.-Y.; Xue, S.-T.; Li, X.; Liu, Z.-Y.; He, X.-B.; Jiang, J.-D.; Zhang, Y.-Q.; Si, S.-Y.; et al. Substituted Benzothiophene or Benzofuran Derivatives as a Novel Class of Bone Morphogenetic Protein-2 Up-Regulators: Synthesis, Structure−Activity Relationships, and Preventive Bone Loss Efficacies in Senescence Accelerated Mice (SAMP6) and Ovariectomized Rats. J. Med. Chem. 2010, 53, 1819–1829. [Google Scholar] [CrossRef]
  19. Xue, S.-T.; Guo, H.-F.; Liu, M.-J.; Jin, J.; Ju, D.-H.; Liu, Z.-Y.; Li, Z.-R. Synthesis of a novel class of substituted benzothiophene or benzofuran derivatives as BMP-2 up-regulators and evaluation of the BMP-2-up-regulating effects in vitro and the effects on glucocorticoid-induced osteoporosis in rats. Eur. J. Med. Chem. 2015, 96, 151–161. [Google Scholar] [CrossRef]
  20. ElHady, A.K.; El-Gamil, D.S.; Chen, P.J.; Hwang, T.L.; Abadi, A.H.; Abdel-Halim, M.; Engel, M. 5-Methoxybenzothiophene-2-Carboxamides as Inhibitors of Clk1/4: Optimization of Selectivity and Cellular Potency. Molecules 2021, 26, 1001. [Google Scholar] [CrossRef]
  21. Eldehna, W.M.; Maklad, R.M.; Almahli, H.; Al-Warhi, T.; Elkaeed, E.B.; Abourehab, M.A.S.; Abdel-Aziz, H.A.; El Kerdawy, A.M. Identification of 3-(piperazinylmethyl)benzofuran derivatives as novel type II CDK2 inhibitors: Design, synthesis, biological evaluation, and in silico insights. J. Enzym. Inhib. Med. Chem. 2022, 37, 1227–1240. [Google Scholar] [CrossRef] [PubMed]
  22. Wadood, A.; Jamal, S.B.; Riaz, M.; Mir, A. Computational analysis of benzofuran-2-carboxlic acids as potent Pim-1 kinase inhibitors. Pharm. Biol. 2014, 52, 1170–1178. [Google Scholar] [CrossRef] [PubMed]
  23. Gao, F.; Wang, T.; Gao, M.; Zhang, X.; Liu, Z.; Zhao, S.-J.; Lv, Z.-S.; Xiao, J. Benzofuran-isatin-imine hybrids tethered via different length alkyl linkers: Design, synthesis and in vitro evaluation of anti-tubercular and anti-bacterial activities as well as cytotoxicity. Eur. J. Med. Chem. 2019, 165, 323–331. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Z.; Liu, Y.; Zhang, J.; Ullah, S.; Kang, N.; Zhao, Y.; Zhou, H. Benzothiophene-2-carboxamide derivatives as SENPs inhibitors with selectivity within SENPs family. Eur. J. Med. Chem. 2020, 204, 112553. [Google Scholar] [CrossRef] [PubMed]
  25. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [Google Scholar] [CrossRef]
  26. Veber, D.F.; Johnson, S.R.; Cheng, H.-Y.; Smith, B.R.; Ward, K.W.; Kopple, K.D. Molecular Properties That Influence the Oral Bioavailability of Drug Candidates. J. Med. Chem. 2002, 45, 2615–2623. [Google Scholar] [CrossRef]
  27. El-Metwally, S.A.; Abou-El-Regal, M.M.; Eissa, I.H.; Mehany, A.B.; Mahdy, H.A.; Elkady, H.; Elwan, A.; Elkaeed, E.B. Discovery of thieno [2, 3-d] pyrimidine-based derivatives as potent VEGFR-2 kinase inhibitors and anti-cancer agents. Bioorganic Chem. 2021, 112, 104947. [Google Scholar] [CrossRef]
  28. Alsaif, N.A.; Dahab, M.A.; Alanazi, M.M.; Obaidullah, A.J.; Al-Mehizia, A.A.; Alanazi, M.M.; Aldawas, S.; Mahdy, H.A.; Elkady, H. New quinoxaline derivatives as VEGFR-2 inhibitors with anticancer and apoptotic activity: Design, molecular modeling, and synthesis. Bioorg. Chem. 2021, 110, 104807. [Google Scholar] [CrossRef]
  29. Alanazi, M.M.; Elkady, H.; Alsaif, N.A.; Obaidullah, A.J.; Alkahtani, H.M.; Alanazi, M.M.; Alharbi, M.A.; Eissa, I.H.; Dahab, M.A. New quinoxaline-based VEGFR-2 inhibitors: Design, synthesis, and antiproliferative evaluation with in silico docking, ADMET, toxicity, and DFT studies. RSC Adv. 2021, 11, 30315–30328. [Google Scholar] [CrossRef]
  30. Abdallah, A.E.; Alesawy, M.S.; Eissa, S.I.; El-Fakharany, E.M.; Kalaba, M.H.; Sharaf, M.H.; Shama, N.M.A.; Mahmoud, S.H.; Mostafa, A.; Al-Karmalawy, A.A.; et al. Design and synthesis of new 4-(2-nitrophenoxy)benzamide derivatives as potential antiviral agents: Molecular modeling and in vitro antiviral screening. New J. Chem. 2021, 45, 16557–16571. [Google Scholar] [CrossRef]
  31. Alanazi, M.M.; Eissa, I.H.; Alsaif, N.A.; Obaidullah, A.J.; Alanazi, W.A.; Alasmari, A.F.; Albassam, H.; Elkady, H.; Elwan, A. Design, synthesis, docking, ADMET studies, and anticancer evaluation of new 3-methylquinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers. J. Enzym. Inhib. Med. Chem. 2021, 36, 1760–1782. [Google Scholar] [CrossRef] [PubMed]
  32. Alsaif, N.A.; Taghour, M.S.; Alanazi, M.M.; Obaidullah, A.J.; Al-Mehizia, A.A.; Alanazi, M.M.; Aldawas, S.; Elwan, A.; Elkady, H. Discovery of new VEGFR-2 inhibitors based on bis ([1, 2, 4] triazolo)[4, 3-a: 3’, 4’-c] quinoxaline derivatives as anticancer agents and apoptosis inducers. J. Enzym. Inhib. Med. Chem. 2021, 36, 1093–1114. [Google Scholar] [CrossRef] [PubMed]
  33. Xia, X.; Maliski, E.G.; Gallant, A.P.; Rogers, D. Classification of Kinase Inhibitors Using a Bayesian Model. J. Med. Chem. 2004, 47, 4463–4470. [Google Scholar] [CrossRef]
  34. BIOVIA, QSAR, ADMET and Predictive Toxicology. Available online: https://www.3dsbiovia.com/products/collaborative-science/biovia-discovery-studio/qsar-admet-and-predictive-toxicology.html.
  35. Molecular Operating Environment (MOE), 2014.09, Chemical Computing Group Inc., Montréal, Canada. Available online: https://www.chemcomp.com.
  36. Mehany, A.B.M.; Belal, A.; Mohamed, A.F.; Shaaban, S.; Abdelhamid, G. Apoptotic and anti-angiogenic effects of propolis against human bladder cancer: Molecular docking and in vitro screening. Biomarkers 2022, 27, 138–150. [Google Scholar] [CrossRef] [PubMed]
  37. Belal, A. 3D-Pharmacophore Modeling, Molecular Docking, and Virtual Screening for Discovery of Novel CDK4/6 Selective Inhibitors. Russ. J. Bioorg. Chem. 2021, 47, 317–333. [Google Scholar] [CrossRef]
  38. Zhaorigetu, I.M.F.; Belal, A.; al Badawi, M.H.; Abdelhady, A.A.; Galala, F.M.A.; El-Sharkawy, A.; El-Dahshan, A.A.; Mehany, A.B. Antiproliferative, Apoptotic Effects and Suppression of Oxidative Stress of Quercetin against Induced Toxicity in Lung Cancer Cells of Rats: In vitro and In vivo Study. J. Cancer 2021, 12, 5249. [Google Scholar] [CrossRef]
  39. Belal, A. Pyrrolizines as Potential Anticancer Agents: Design, Synthesis, Caspase-3 activation and Micronucleus (MN) Induction. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem.-Anti-Cancer Agents) 2018, 18, 2124–2130. [Google Scholar] [CrossRef]
  40. Belal, A. Synthesis, molecular docking and antitumor activity of novel pyrrolizines with potential as EGFR-TK inhibitors. Bioorg. Chem. 2015, 59, 124–129. [Google Scholar] [CrossRef]
  41. El-Helby, A.-G.A.; Ayyad, R.R.; El-Adl, K.; Elkady, H. Phthalazine-1, 4-dione derivatives as non-competitive AMPA receptor antagonists: Design, synthesis, anticonvulsant evaluation, ADMET profile and molecular docking. Mol. Divers. 2019, 23, 283–298. [Google Scholar] [CrossRef]
  42. El-Zahabi, M.A.; Elbendary, E.R.; Bamanie, F.H.; Radwan, M.F.; Ghareib, S.A.; Eissa, I.H. Design, synthesis, molecular modeling and anti-hyperglycemic evaluation of phthalimide-sulfonylurea hybrids as PPARγ and SUR agonists. Bioorg. Chem. 2019, 91, 103115. [Google Scholar] [CrossRef]
  43. Ibrahim, M.K.; Eissa, I.H.; Alesawy, M.S.; Metwaly, A.M.; Radwan, M.M.; ElSohly, M.A. Design, synthesis, molecular modeling and anti-hyperglycemic evaluation of quinazolin-4 (3H)-one derivatives as potential PPARγ and SUR agonists. Bioorg. Med. Chem. 2017, 25, 4723–4744. [Google Scholar] [CrossRef] [PubMed]
  44. El-Demerdash, A.; Metwaly, A.M.; Hassan, A.; El-Aziz, A.; Mohamed, T.; Elkaeed, E.B.; Eissa, I.H.; Arafa, R.K.; Stockand, J.D. Comprehensive virtual screening of the antiviral potentialities of marine polycyclic guanidine alkaloids against SARS-CoV-2 (COVID-19). Biomolecules 2021, 11, 460. [Google Scholar] [CrossRef] [PubMed]
  45. El-Gamal, K.M.; El-Morsy, A.M.; Saad, A.M.; Eissa, I.H.; Alswah, M. Synthesis, docking, QSAR, ADMET and antimicrobial evaluation of new quinoline-3-carbonitrile derivatives as potential DNA-gyrase inhibitors. J. Mol. Struct. 2018, 1166, 15–33. [Google Scholar] [CrossRef]
  46. Alanazi, M.M.; Mahdy, H.A.; Alsaif, N.A.; Obaidullah, A.J.; Alkahtani, H.M.; Al-Mehizia, A.A.; Alsubaie, S.M.; Dahab, M.A.; Eissa, I.H. New bis ([1, 2, 4] triazolo)[4, 3-a: 3′, 4′-c] quinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers: Design, synthesis, in silico studies, and anticancer evaluation. Bioorg. Chem. 2021, 112, 104949. [Google Scholar] [CrossRef]
  47. Eissa, I.H.; El-Helby, A.G.A.; Mahdy, H.A.; Khalifa, M.M.; Elnagar, H.A.; Mehany, A.B.; Metwaly, A.M.; Elhendawy, M.A.; Radwan, M.M.; ElSohly, M.A.; et al. Discovery of new quinazolin-4 (3H)-ones as VEGFR-2 inhibitors: Design, synthesis, and anti-proliferative evaluation. Bioorg. Chem. 2020, 105, 104380. [Google Scholar] [CrossRef]
  48. Eissa, I.H.; Dahab, M.A.; Ibrahim, M.K.; Alsaif, N.A.; Alanazi, A.; Eissa, S.I.; Belal, A.; Beauchemin, A.M. Design and discovery of new antiproliferative 1, 2, 4-triazin-3 (2H)-ones as tubulin polymerization inhibitors targeting colchicine binding site. Bioorg. Chem. 2021, 112, 104965. [Google Scholar] [CrossRef]
  49. Eissa, I.H.; Ibrahim, M.K.; Metwaly, A.M.; Belal, A.; Mehany, A.B.; Abdelhady, A.A.; Elhendawy, M.A.; Radwan, M.M.; ElSohly, M.A.; Mahdy, H.A.J.B.C. Design, molecular docking, in vitro, and in vivo studies of new quinazolin-4 (3H)-ones as VEGFR-2 inhibitors with potential activity against hepatocellular carcinoma. Bioorg. Chem. 2021, 107, 104532. [Google Scholar] [CrossRef] [PubMed]
  50. Mehany, A.B.M.; Belal, A.; Santali, E.Y.; Shaaban, S.; Abourehab, M.A.S.; El-Feky, O.A.; Diab, M.; Galala, F.M.A.A.; Elkaeed, E.B.; Abdelhamid, G. Biological Effect of Quercetin in Repairing Brain Damage and Cerebral Changes in Rats: Molecular Docking and In Vivo Studies. BioMed Res. Int. 2022, 2022, 1–12. [Google Scholar] [CrossRef]
  51. Belal, A.; Elanany, M.A.; Raafat, M.; Hamza, H.T.; Mehany, A.B.M. Calendula officinalis Phytochemicals for the Treatment of Wounds Through Matrix Metalloproteinases-8 and 9 (MMP-8 and MMP-9): In Silico Approach. Nat. Prod. Commun. 2022, 17, 1934578X221098848. [Google Scholar] [CrossRef]
  52. Sun, X.; Belal, A.; Elanany, M.A.; Alsantali, R.I.; Alrooqi, M.M.; Mohamed, A.R.; Hasabelnaby, S. Identification of Some Promising Heterocycles Useful in Treatment of Allergic Rhinitis: Virtual Screening, Pharmacophore Mapping, Molecular Docking, and Molecular Dynamics. Russ. J. Bioorg. Chem. 2022, 48, 438–456. [Google Scholar]
  53. Belal, A.; Gawad, N.M.A.; Mehany, A.B.M.; Abourehab, M.A.S.; Elkady, H.; Al-Karmalawy, A.A.; Ismael, A.S. Design, synthesis and molecular docking of new fused 1H-pyrroles, pyrrolo[3,2-d]pyrimidines and pyrrolo[3,2-e][1, 4]diazepine de-rivatives as potent EGFR/CDK2 inhibitors. J. Enzym. Inhib. Med. Chem. 2022, 37, 1884–1902. [Google Scholar] [CrossRef] [PubMed]
  54. Elkaeed, E.B.; Elkady, H.; Belal, A.; Alsfouk, B.A.; Ibrahim, T.H.; Abdelmoaty, M.; Arafa, R.K.; Metwaly, A.M.; Eissa, H.I. Mul-ti-Phase In Silico Discovery of Potential SARS-CoV-2 RNA-Dependent RNA Polymerase Inhibitors among 3009 Clinical and FDA-Approved Related Drugs. Processes 2022, 10, 530. [Google Scholar] [CrossRef]
  55. El-Naggar, A.M.; Eissa, I.H.; Belal, A.; El-Sayed, A.A. Design, eco-friendly synthesis, molecular modeling and anticancer eval-uation of thiazol-5 (4 H)-ones as potential tubulin polymerization inhibitors targeting the colchicine binding site. RSC Adv. 2020, 10, 2791–2811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Reported benzothiophene and benzofuran derivatives as BMP-2 up-regulators. a UPR: Up-regulatory percent Activities on BMP-2 expression.
Figure 1. Reported benzothiophene and benzofuran derivatives as BMP-2 up-regulators. a UPR: Up-regulatory percent Activities on BMP-2 expression.
Molecules 27 05571 g001
Figure 2. Reported benzothiophenes as CIK1,2,4 inhibitors (V), raloxifene (VI): anti-breast cancer, benzofuran derivatives VII and VIII as CDK2 and Pim1 inhibitors, respectively.
Figure 2. Reported benzothiophenes as CIK1,2,4 inhibitors (V), raloxifene (VI): anti-breast cancer, benzofuran derivatives VII and VIII as CDK2 and Pim1 inhibitors, respectively.
Molecules 27 05571 g002
Figure 3. Compounds 11, 26, 27, 30, 31, 34, 35, 65, and 72 exhibited medium to very low BBB penetration levels.
Figure 3. Compounds 11, 26, 27, 30, 31, 34, 35, 65, and 72 exhibited medium to very low BBB penetration levels.
Molecules 27 05571 g003
Figure 4. (A) The 3D and (B) 2D binding of co-crystallized ligand in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 4. (A) The 3D and (B) 2D binding of co-crystallized ligand in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g004aMolecules 27 05571 g004b
Figure 5. (A) The 3D and (B) 2D binding of 26 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 5. (A) The 3D and (B) 2D binding of 26 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g005aMolecules 27 05571 g005b
Figure 6. (A) The 3D and (B) 2D binding of 27 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 6. (A) The 3D and (B) 2D binding of 27 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g006aMolecules 27 05571 g006b
Figure 7. (A) The 3D and (B) 2D binding of 31 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 7. (A) The 3D and (B) 2D binding of 31 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g007aMolecules 27 05571 g007b
Figure 8. (A) The 3D and (B) 2D binding of 35 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 8. (A) The 3D and (B) 2D binding of 35 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g008aMolecules 27 05571 g008b
Figure 9. (A) The 3D and (B) 2D binding of 72 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Figure 9. (A) The 3D and (B) 2D binding of 72 in the active site of BMP-2-inducible kinase (hydrogen bonds = green dashed lines, electrostatic interactions = orange dashed lines, pi-pi interactions = purple dashed lines, and pi-alkyl interactions = light pink dashed lines).
Molecules 27 05571 g009aMolecules 27 05571 g009b
Figure 10. The 2D binding of compound 11 with Cell division protein kinase 2 (PDB ID: 3lfn).
Figure 10. The 2D binding of compound 11 with Cell division protein kinase 2 (PDB ID: 3lfn).
Molecules 27 05571 g010
Figure 11. The 2D binding of compound 26 with Casein kinase II subunit alpha (PDB ID: 5csp).
Figure 11. The 2D binding of compound 26 with Casein kinase II subunit alpha (PDB ID: 5csp).
Molecules 27 05571 g011
Figure 12. The 2D binding of compound 30 with cell division protein kinase 2 (PDB ID: 2wev).
Figure 12. The 2D binding of compound 30 with cell division protein kinase 2 (PDB ID: 2wev).
Molecules 27 05571 g012
Figure 13. The 2D binding of compound 35 with Eukaryotic translation initiation factor 2-alpha kinase 3 (PDB ID: 4 × 7 h).
Figure 13. The 2D binding of compound 35 with Eukaryotic translation initiation factor 2-alpha kinase 3 (PDB ID: 4 × 7 h).
Molecules 27 05571 g013
Table 1. Physicochemical properties of the tested compounds passed Lipinski’s and Veber’s Rules.
Table 1. Physicochemical properties of the tested compounds passed Lipinski’s and Veber’s Rules.
Comp. Lipinski’s RuleVeber’s Rule
Num HDNum HAM WtAlogPNum Rotatable BondsTPSA
1108498.5274.9941090.57
2613208.2342.596274.77
2735251.2581.454117.86
2813284.334.18474.77
2916329.3274.0745120.59
3016329.3274.0745120.59
3116329.3274.0745120.59
3213298.3564.666474.77
3325416.4924.543695.67
3447459.5173.3968138.76
3525492.5886.126895.67
4034388.4824.595692.59
6136481.5665.5118111.46
6226482.555.5218108.81
6325498.6166.088123.91
6526493.5764.9768108.56
6825430.5194.462795.67
6925444.5455.129895.67
7025458.5725.585995.67
7125458.5725.591895.67
7226499.6044.9298136.8
Table 2. Predicted computational ADMET for the tested candidate.
Table 2. Predicted computational ADMET for the tested candidate.
Comp.BBB Level aSolubility Level bAbsorption Level cCYP2D6 Prediction dPPB Prediction e
11421falseTrue
26230falseTrue
27330falseTrue
28120falseTrue
29220falseTrue
30220falsetrue
31220falseTrue
32120falseTrue
33120falseTrue
34420falseTrue
35411falseTrue
40120falseTrue
61411falseTrue
62411falseTrue
63411falseTrue
65420falseTrue
68120falseTrue
69120falseTrue
70110falseTrue
71111falseTrue
72220falseTrue
IDK420falseTrue
a BBB level, blood–brain barrier level, 0 = very high, 1 = high, 2 = medium, 3 = low, 4 = very low. b Solubility level, 1 = very low, 2 = low, 3 = good, 4 = optimal. c Absorption level, 0 = good, 1 = moderate, 2 = poor, 3 = very poor. d CYP2D6, cytochrome P2D6, TRUE = inhibitor, FALSE = non inhibitor. e PBB, plasma protein binding, FALSE means less than 90%, TRUE means more than 90%.
Table 3. Calculated ∆G, amino acid interactions, and distances for the best five candidates and co-crystallized ligand (IDK) against BMP-2-inducible kinase (∆G in Kcal/mol).
Table 3. Calculated ∆G, amino acid interactions, and distances for the best five candidates and co-crystallized ligand (IDK) against BMP-2-inducible kinase (∆G in Kcal/mol).
Comp∆G
Kcal/mol
No. of H BondsDistance (Å)Amino Acid Involved
26−22.9042.59Glu131
2.15Cys133
2.92Cys197
2.78Gln137
27−23.3442.92Cys133
2.39Cys197
2.35, 2.44Gln137
31−22.2242.24Glu131
3.08, 3.18Cys133
2.52Cys197
35−24.2552.80, 2.72Asn140
2.33Cys 133
2.85Gln 137
2.86Leu 57
72−24.2252.72Glu131
2.75Glu184
2.68Lys 182
2.76Tyr 132
2.65Gly 60
Co-crystallized ligand (IDK)−22.7253.22Glu131
2.79, 2.83Cys133
3.36Asn185
2.89Gln137
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Belal, A.; Elkady, H.; Al-Karmalawy, A.A.; Amin, A.H.; Ghoneim, M.M.; El-Sherbiny, M.; Al-Serwi, R.H.; Abdou, M.A.; Ibrahim, M.H.; Mehany, A.B.M. Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations. Molecules 2022, 27, 5571. https://doi.org/10.3390/molecules27175571

AMA Style

Belal A, Elkady H, Al-Karmalawy AA, Amin AH, Ghoneim MM, El-Sherbiny M, Al-Serwi RH, Abdou MA, Ibrahim MH, Mehany ABM. Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations. Molecules. 2022; 27(17):5571. https://doi.org/10.3390/molecules27175571

Chicago/Turabian Style

Belal, Amany, Hazem Elkady, Ahmed A. Al-Karmalawy, Ali H. Amin, Mohammed M. Ghoneim, Mohamed El-Sherbiny, Rasha Hamed Al-Serwi, Mohamed Attia Abdou, Mona H. Ibrahim, and Ahmed B. M. Mehany. 2022. "Discovery of Some Heterocyclic Molecules as Bone Morphogenetic Protein 2 (BMP-2)-Inducible Kinase Inhibitors: Virtual Screening, ADME Properties, and Molecular Docking Simulations" Molecules 27, no. 17: 5571. https://doi.org/10.3390/molecules27175571

Article Metrics

Back to TopTop