Next Article in Journal
Special Issue “Recent Advances in Oral Drug Delivery Development”
Previous Article in Journal
Analgesic Efficacy and Safety of Tapentadol Immediate Release in Bunionectomy: A Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Imidazopyridine-Based Thiazole Derivatives as Potential Antidiabetic Agents: Synthesis, In Vitro Bioactivity, and In Silico Molecular Modeling Approach

1
Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
2
Department of Chemistry, COMSATS University Islamabad, Islamabad 45550, Pakistan
3
Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
4
Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(9), 1288; https://doi.org/10.3390/ph16091288
Submission received: 3 August 2023 / Revised: 5 September 2023 / Accepted: 7 September 2023 / Published: 13 September 2023
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
A new series of thiazole derivatives (4a-p) incorporating imidazopyridine moiety was synthesized and assessed for their in vitro potential α-glucosidase potency using acarbose as a reference drug. The obtained results suggested that compounds 4a (docking score = −13.45), 4g (docking score = −12.87), 4o (docking score = −12.15), and 4p (docking score = −11.25) remarkably showed superior activity against the targeted α-glucosidase enzyme, with IC50 values of 5.57 ± 3.45, 8.85 ± 2.18, 7.16 ± 1.40, and 10.48 ± 2.20, respectively. Upon further investigation of the binding mode of the interactions by the most active scaffolds with the α-glucosidase active sites, the docking analysis was accomplished in order to explore the active cavity of the α-glucosidase enzyme. The interpretation of the results showed clearly that scaffolds 4a and 4o emerged as the most potent α-glucosidase inhibitors, with promising excellent binding interactions with the active site of the α-glucosidase enzyme. Furthermore, utilizing a variety of spectroscopic methods, such as 1H-NMR, 13C-NMR, and HREI-MS, the precise structures of the synthesized scaffolds were determined.

1. Introduction

Diabetes mellitus is characterized as a metabolic condition that has numerous non-infectious etiologies. It is described as persistent hyperglycemia and conflicts between the carbohydrate, protein, and fat metabolisms as a result of abnormalities in the secretion of insulin or its action, or both [1]. To avoid the consequences of diabetes, including peripheral arteriopathy, retinopathy, heart coronary disease, nephropathy, and neuropathy, blood glucose levels must be normalized, especially postprandial hyperglycemia [2]. In numerous modern and conventional medical systems around the world, a wide range of synthetic and natural compounds have been identified as potential sources of anti-diabetic special effects. Many of them have a reputation for being excellent diabetes treatments [3]. Many medications used to treat type-2 diabetes today not only have side effects but also have limited tolerability and efficacy and are not frequently successful in maintaining normal glucose levels in the blood [4,5]. The primary job of α-glucosidase (a member of the glycosyl hydrolases) is to hydrolyze the non-reducing terminal, α-D-glucosidase (1,4-linked), to facilitate the α-D-glucose discharge. The α-glucosidase enzyme has attracted particular attention among pharmaceutical and medicinal chemists, as it was found in earlier investigations that inhibiting its enzymatic potential delayed glucose absorption and decreased postprandial blood glucose levels. The inhibitors of α-glucosidase are crucial in preventing postprandial hyperglycemia and delaying or inhibiting the digestion and absorption of carbohydrates [6]. As a result, they have a wide range of applications, many of which may be helpful in type-2 diabetes treatment. Due to their distinct biological characteristics, heterocyclic molecules are of particular attention to chemists (medicinal). The most prevalent and essential analogs that exist exclusively in numerous pharmaceutical drugs, natural bioactive materials, and natural products are heterocyclic molecules that contain nitrogen [7]. The design and development of synthesis methods and novel technologies have been sparked by the discovery that N-heterocycles are a crucial structural entity in a number of physiologically potent compounds. Thiazole is a component of about 200 naturally occurring alkaloids that have been found thus far in various plants, microbes, and animals [8]. Among N-containing heterocyclic compounds, numerous substituted thiazoles have been produced in an effort to uncover more potential thiazole-based drugs [9]. The thiazole core skeleton-containing derivatives have been reported to show significant biological activities such as anti-diuretic, anti-Alzheimer, and anti-bacterial activities [10]. The hybrid analogs based on the thiazole moiety are also used as pharmacologically intriguing derivatives and have been shown to exhibit interesting biological effects, including those that have anti-inflammatory [11], anti-microbial [12,13], anti-hypoxic [14], analgesic [15,16], anti-cancer [17,18], anti-asthmatic [19,20], and anti-hypertensive activities [21]. Owing to the broad spectrum of the biological profile of the thiazole ring, it was also discovered that many commercially available biologically active medications, including ruvaconazole, abafungin, vorelaxin, tiabendazole, ruvaconazole, tiazofurin, aztreonam, and azereonam have the thiazole skeleton in their structures (Figure 1) [22,23].
Additionally, essential for the study of their biological and pharmaceutical applications, the N-containing heterocyclic compounds have gained much attention [24]. One in particular is imidazopyridine, with rings of both imidazole and pyridine, which constitutes a typical, preferred scaffold, containing antifungal properties and working as an antagonist to the H1 receptor as well as a kinase and anti-lipase inhibitors (Figure 2) [25].
In order to design and synthesize hybrid analogs as potential anti-diabetic agents, the molecular hybridization technique has been widely used. This method mainly involves combining two or more distinct pharmacophore components into a single molecule with the same backbone. These hybrid molecules could be better than standard drugs. The physiologically significant heterocyclic moieties, including imidazopyridine bearing thiazole [26,27], and, as in this study, both thiazole [28,29,30] and imidazopyridine [31,32,33], are currently being combined through molecular hybridization to create new hybrid compounds (Figure 3).As was previously mentioned, the imidazopyridine and thiazole moieties are crucial as potential anti-diabetic agents, so hybrid analogs (4a-4p) with combinations of these two moieties were afforded and then tested for their ability to inhibit alpha-glucosidase in vitro (Figure 3).

2. Results

2.1. Chemistry

In order to synthesize hybrid analogs of imidazopyridine-based thiazole (4a-p), thiosemicarbazide (1) was initially reacted to fluoro-containing imidazopyridine 3-carbaldehyde in methanol through the addition of a few drops of CH3COOH. The resulting mixture was refluxed for 6 hrs over a pre-heated sand bath to afford imidazopyridine-based thiosemicarbazone substrate (2) and further under wentcyclization on stirring with variably 2-bromoacetophenone (3) in Et3N and EtOH. The mixture of reaction was stirred and refluxed until it yielded the final product (TLC, 16 hrs reflux). The solvent was evaporated by employing low pressure to generate a solid residue of targeted analogs and then washed with n-hexane to form imidazopyridine-based thiazole analogs (4a-p) in a good yield (Scheme 1).

2.2. Biological Analysis (4a-p)

α-Glucosidase Inhibitory Activity

The inhibition profile of each synthesized scaffold was shown in vitro. The results were compared with acarbose as a reference. The inhibitory potential of the newly synthesized scaffolds ranged from 5.57 ± 3.45 to 63.46 ± 5.28 μM in comparison to the acarbose reference (IC50 = 48.71 ± 2.65 μM).
Generally, it was summarized based on the aforementioned observation that theα-glucosidase activity of imidazopyridine-based thiazoles (4a-p) was largely affected by the introduction of attached substituents; the nature and location with respect to the phenyl ring can vary. Moreover, the inhibitory potential of synthetic imidazopyridine-based thiazole analogs was also impacted by the change in the number of attached substituent(s) around the aryl parts (Table 1).

2.3. Molecular Docking Study

A molecular docking study has been performed to assess the correlation between in vitro and in silico studies of all the active synthesized 4g, 4a, 4o, and 4p analogs. All of these active compounds subsequently bind effectively in the target’s active site with a variety of binding affinities and also correlate with in vitro studies. All of these active analogs have the same chemistry with slight variations in different positions. Different interactions with the target alpha-glucosidase active sites were shown by these various functional moieties at different positions of active analogs (Figure 4, Figure 5, Figure 6 and Figure 7).

3. Discussion

3.1. Structure–Activity Relationship (SAR) Study for α-Glucosidase Inhibitory Profile

Structure–activity relationship (SAR) studies were conducted for the newly synthesized analogs (4a-p) by bringing substituent(s) around the aryl part, and any variation in inhibitory potentials was due to variation in number, position, and ED/EW nature of the substituent(s) linked to the aryl part while keeping the imidazole and thiazole rings constant (Table 2).
The compounds (4a, 4g, 4o, and 4p) bearing substituent(s) of either strong EWG nature, such as -CF3 and -NO2, or strong H-bond (-OH) making substituent(s) attached to variable positions of the extended aromaticbenzene ring were recognized asdemonstrating significant potentcy, when compared to the remaining analogs of the current synthesized series. The scaffold 4g bearing ortho-CF3 and meta-nitro substituents at the phenyl ring was shown to exhibit many times more potency than the standard acarbose drug, as well as the rest of the analogs of the synthesized series. However, the analog 4a, which also holds the same -CF3 and -NO2 moieties at the phenyl ring, has a different position of the -CF3 moiety around the aryl part and was found somewhat less potent than its counterpart 4g, even through this scaffold 4a was identified as the second-most potent inhibitor of the target among the series. The enhanced inhibitory potentials of both of these scaffolds, 4g and 4a, might be due to the attached substituents (-CF3 and -NO2) having a strong EW nature and are also capable of interacting through side-wise association with the target active sites. Further, the comparison of scaffolds 4g (which holds 2-CF3 and 5-NO2 groups at the phenyl part) with analog 4o (bearing 2-OH and 5-NO2 groups at the aryl part) demonstrated that the 4g scaffold exhibited more enhanced inhibitory potentials than the 4o analog. Both of these scaffolds differ from one another due to the nature of substituents (5g holds 2-CF3, while 4o bears 2-OH group) at the 2-position of the phenyl ring. The better inhibitory potentials of scaffold 4g revealed that the -CF3 moiety established better interactions than the -OH group with target active sites. Similarly, analog 4p (which holds di-hydroxy at the 2,4-position of the phenyl ring) displayed many times more potency than the standard acarbose drug but was found less potent than its counterparts 4a, 4g, and 4o. In addition, the comparison of analog 4o (bearing 2-hydroxy and 5-NO2) with 4p (bearing di-hydroxy at the 2,4-position) shows that analog 4o exhibited enhanced inhibitory potentials owing to the attached EW -NO2 group at the 5-position, which interacts better with the target active sites (Figure 8).
It was revealed by the SAR studies that the attachment of substituent(s) (-CH3 and -OCH3,either alone or in combination) was capable of forming weak interactions with the target active sites and hence resulted in declined potency. Analog 4k, which holds -CH3 at the ortho- and -Cl moieties at the para-position of the aryl part, showed good potency as compared to the standard drug. This better activity of analog 4k was due to the strong e-withdrawing effects of the -Cl moiety, which makes the aryl ring more probed for interaction with target α-glucosidase active sites. Similarly, 4m has lower potency than the standard drug, which might be due to the presence of two -OCH3 groups on the 2,4-position of the aryl part. These two methoxy groups have an electron-donating effect, and due to this, it will lower the inhibition activity. Moreover, if we compare analog 4m, bearing di-OCH3 groups at the 2,4-position of the aryl part, with scaffold 4f, bearing di-CH3 moieties at the 2,4-position of the aryl part, the analog 4f displayed superior activity than that of analog 4m. This difference was due to the fact that the methyl group is the ring activator group which is less electronegative than the oxygen of the methoxy group and a charge is not produced on the aryl ring as compared to the methoxy groups of 4m; therefore, the inhibition activity of 4f is somewhat better than 4m. However, compound 4e holds both the ortho-OCH3 and para-CH3 groups and makes a somewhat better competitor of α-glucosidase activity as compared to the standard drug (Figure 9).
Moreover, the inhibition profiles of the newly afforded scaffolds were found to be encouraging by attachment of -Cl, -NO2, and -OH groups, either alone or in combination, at variable positions of the aryl part. The analog 4b, bearing the-NO2 moiety at the ortho- and the -Cl moieties at the para-position of the phenyl moiety has emerged as the potent inhibitor of the α-glucosidase enzyme (IC50 = 13.63 ± 1.67 μM) in comparison to the reference drug (48.71 ± 2.65). This elevation in activity of scaffold 4b might be owing to the attached -NO2 and -Cl groups having strong e-withdrawing natures. Similarly, analog 4l, bearing di-Cl groups at the 2,4-position of the aryl part, shows two-fold more potency as compared to the standard drug, while showing a slightly lower potency as compared to analog 4b. This better potency was due to the attached -Cl group, which is a good e-withdrawing group that will enhance the potency of the molecule. Moreover, the compound 4j has also shown two-fold more potency as compared to the standard drug due to the presence of an electron-withdrawing group of -NO2 at the 2-position of the phenyl ring. The inhibition profile of scaffold 4i is also good/moderate with MIC value (34.91 ± 5.84) as compared to the standard acarbose (48.71 ± 2.65) drug, due to the presence of -OCH3 and -OH, because the -OCH3 group also increases the inhibitory potential to some extent, as well as the -OH holds the tendency of making an H-bond with the target active sites and therefore increases the potency of the molecule. This difference found in the inhibition profiles of both scaffolds 4b and 4l might be due to the stronger EW nature possessed by -NO2 than that of the -Cl moiety, which holds a weaker EW effect, as well as the fact that the-NO2 groups also have a tendency to established strong side-wise interactions by involving its oxygen with the target active sites and hence, enhanced the enzymatic potentials when the results are compared to the reference drug (Figure 10).
Nonetheless, the SAR studies suggested that the incorporation of groups of a bulky nature, such as -Ph, -toluene, and -N(CH3)2 moieties, at the para-position of the aryl part, resulted in a decrease in inhibitory potentials against the targeted alpha-glucosidase enzyme. Analog 4c, bearing para-phenyl moiety at the aryl part, and analog 4n, having p-tolyl moiety at the aryl part, emerged as the least potent competitors of the alpha-glucosidase enzyme among the series. This decreased inhibitory potential of both analogs 4c and 4n might be due to the bulky nature of attached substituent(s). Apart from this, compounds 4d (holds -N(CH3)2 at the para-position) and 4h (holds -CH3 at the ortho-position of the aryl part) showed moderately good results when compared with the standard acarbose drug. Hence, 4c and 4n showed reduced potency comparable with the standard drug, while compounds 4d and 4n showed moderately good potency as compared with the acarbose drug (Figure 11).

3.2. Molecular Docking Study

3.2.1. Discussion

Through in vitro analysis, compound 4g was found to be the most active. The targeted active analog sites interacted with the most active analog. The most potent analog’s detailed interactions reveal that this scaffold provided several significant interactions with the active α-glucosidase target sites, including Phe200 (pi-pi T shaped), Glu595 (pi-anion), Val96 (CHB, pi-alkyl and pi-pi stacked), Phe51 (pi-alkyl), Thr175 (carbon hydrogen bond), His94 (halogen (fluorine)), Phe95 (halogen (fluorine) and pi-alkyl), and Arg91 (pi-alkyl) interactions.
The PLI of the second-most effective scaffold 4a showed that, despite a structural similarity with the most active analog 4g, the two analogs differ only in the location of the -CF3 moiety in analog 4a; it is attached at the meta-position of the aryl ring, whereas in analog 4g, it is attached at the ortho-position of the aryl part of the thiazole. The different positions of the functional moiety -CF3 around the aryl part may be the cause of the reduction in the enzymatic potential, which makes both the active 4g and 4a analogs interact differently. The detailed PLI of the second-most active scaffold 4a showed that this analog adopted various noteworthy interactions with the target α-glucosidase active sites, including Trp90 (pi-sulfur and pi-pi T shaped), Arg91 (pi-pi stacked), Gln202 (CHB), His94 (halogen (fluorine) and pi-pi stacked), Val96 (pi-alkyl), Val201 (pi-alkyl), and Phe200 (halogen (fluorine) and pi-alkyl) interactions.
According to the docking study, substituents that form strong hydrogen bonds with the target active sites also enhance the inhibition profile; as a result, analog 4o (bearing ortho-hydroxy and meta-nitro moieties at the aryl part) established various important interactions with the target α-glucosidase active sites, such as Phe51 (pi-pi T shaped), Arg91 (pi-pi stacked), Val96 (pi-pi stacked and pi-alkyl), Gln202 (CHB), Glu595 (pi-anion), Phe95 (carbon–hydrogen bond), and His95 (pi-pi T shaped) interactions.
As an additional active inhibitor of the α-glucosidase enzyme, the analog 4p with di-hydroxy substituents at the 2,4-position of the aryl part of the thiazole ring showed various significant important interactions, such as Arg91 (pi-alkyl), Phe200 (pi-pi T shaped), Val96 (pi-alkyl), Val201 (pi-alkyl), Glu595 (pi-anion), His94 (halogen (fluorine) and carbon–hydrogen bond), and Phe95 (halogen (fluorine)) interactions.

3.2.2. ADME Analysis

An ADME analysis was performed using the online software Swiss ADME in order to explore the better potentials of synthesized compounds having drug-likeness properties. The subjected analogs 4g, 4a, 4o, and 4p were identified by similar criteria, which showed varied drug-like properties as shown in Graph 1, Graph 2, Graph 3, Graph 4.

4. Materials and Methods

4.1. General Information

Using an electro-thermal melting point apparatus called the Stuart-SMP30 in open glass capillaries, the uncorrected melting points (°C) were ascertained. Nuclear magnetic resonance (NMR) spectra for 1H and 13C were obtained on a JEOL ECA-500 II NMR spectrometer at 600 MHz and 150 MHz, respectively. The chemical shifts represented in ppm downfield from the internal standard, tetramethylsilane, and the coupling constants (J) are given in Hz. Deuteriodimethyl sulfoxide was used as the solvent (DMSO-d6). The reactions were seen and the purity of the finished products was evaluated using thin-layer chromatography, utilizing silica-gel-precoated aluminum sheets (60 F254, Merck) and visualization with ultraviolet light (UV) at 365 and 254 nm.

4.2. General Procedure for Synthesis of Imidazopyridine-Based Thiazole Analogs (4a-p)

The synthesis of imidazopyridine-based thiazole was completed in two steps: In the first step, imidazopyridine-3-carbaldehyde (1 equivalent) was added to thiosemicarbazide (1) (1 equivalent) solution being stirred in methanol (10 mL) and CH3COOH (catalyst). The residue was refluxed for 6 hrs to deliver imidazopyridine-based thiosemicarbazone (2). In the last step, an intermediate (2) (1 equivalent) was subjected to cyclization with different substituted phenacyl bromide (3) (1 equivalent) in methanol (10 mL) and triethylamine (catalyst) to afford the targeted imidazopyridine-based thiazole analogs (4a-p).

4.3. Spectral Analysis (Provided in Supplementary Information)

4.3.1. Assay Protocol for α-Glucosidase Inhibition

With slight modification to an earlier technique, the α-glucosidase inhibition profiles of the synthesized scaffolds were assessed spectrophotometrically [34]. Amounts of 10 μL of the sample, 120 μL of 100 mM potassium phosphate buffer, and 20 mL of a 0.5 U/mL α-glucosidase solution (0.3 mM, in buffer) were combined for each chemical. As an inhibitor, acarbose was commonly employed. Each combination was incubated at 37 °C for a separate period of 15 min. An amount of 20 μL of the substrate 4-nitrophenyl—D-α-glucopyranoside (pNPG) (5 mM) was added to the solution to start the enzymatic reaction. Once more, the mixture was incubated for 15 min at 37 °C. At that point, 80 μL of a 0.2 M sodium carbonate solution was administered to terminate the process. The mixture that included no enzymes also functioned as a blank sample. At 405 nm, the absorbance was finally calculated. To accurately measure the background absorbance, the blank sample, which replaced the substrate with 50 μL of the buffer, was examined. Instead of test samples (acarbose), the positive control sample was composed of 10 L of DMSO (dimethyl sulfoxide). Assuming that A represents the absorbance of the control without test samples and B represents the absorbance with test samples present, the percentage of enzyme inhibition was computed as (1 B/A) 100. The analyses were completed in triplicate, and the mean SD of the outcomes was provided.

4.3.2. Assay Protocol for Molecular Docking Study

To determine the binding affinities between newly accessible active scaffolds and the target α-glucosidase receptor active sites, docking investigations were carried out using the AutoDock Tools 1.5.6. on the Discovery Studio R2 64-bit Client system; interactive molecular structure visualization and analysis were performed. The RCSB Protein Data Bank (PDB) provided the α-glucosidase three-dimensional (3D) structure. The chemical structures of the ligands were obtained from the Pub Chem compound database. Using Open Babel, the PDB files were converted into PDBQT files. For docking analysis, the ligand and target PDB coordinates were optimized. The geometry was optimized using Gaussian 09 with the addition of the missing residues. These coordinates had the lowest energy and were the most stable. Further details were given in previously reported work [35,36].

5. Conclusions

In summary, a new class of functionalized imidazopyridine-bearing thiazole analogs was synthesized, and its α-glucosidase potency was exhibited. The spectral analysis, including 1H-NMR, 13C-NMR, and HREI-MS, confirmed the structures of the newly synthesized compounds. The in vitro α-glucosidase study against acarbose revealed that four newly synthesized analogs, 4a, 4g, 4o, and 4p, (bearing substituents either capable of forming hydrogen bonds or having strong EW natures) showed α-glucosidase activity with minimum IC50 values. Additionally, molecular docking studies revealed that these active analogs occupied the protein-binding pockets of α-glucosidase with strong binding interactions.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph16091288/s1.

Author Contributions

Conceptualization, W.R.; methodology, R.H.; validation, A.M.; formal analysis, S.K. and L.R.; investigation, Y.K.; resources, A.S.A.; data curation, M.H.; writing—original draft preparation, W.R.; writing—review and editing, L.R.; supervision, W.R.; funding acquisition, A.S.A. All authors have read and agreed to the published version of the manuscript.

Funding

The authors extend their appreciation to the Researchers Supporting Project number (RSPD2023R754), King Saud University, Riyadh, Saudi Arabia for funding this research. The authors also extend their appreciation to Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2023R342), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia for funding this research.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Association, A.D. 5. Prevention or delay of type 2 diabetes: Standards of medical care in diabetes-2018. Diabetes Care 2018, 41, S51–S54. [Google Scholar] [CrossRef]
  2. Javid, M.T.; Rahim, F.; Taha, M.; Rehman, H.U.; Nawaz, M.; Imran, S.; Uddin, I.; Mosaddik, A.; Khan, K.M. Synthesis, in vitro α-glucosidase inhibitory potential and molecular docking study of thiadiazole analogs. Bioorg. Chem. 2018, 78, 201–209. [Google Scholar] [CrossRef]
  3. Swaroopa, P.; Code, Q. Review on antidiabetic activity on medicinal plants. Int. J. Pharmacol. Res. 2017, 7, 230–235. [Google Scholar]
  4. Xu, X.; Wang, G.; Zhou, T.; Chen, L.; Chen, J.; Shen, X. Novel approaches to drug discovery for the treatment of type 2 diabetes. Expert Opin. Drug Discov. 2014, 9, 1047–1058. [Google Scholar] [CrossRef]
  5. Prabhakar, P.K.; Doble, M. Mechanism of action of natural products used in the treatment of diabetes mellitus. Chin. J. Integr. Med. 2011, 17, 563–574. [Google Scholar] [CrossRef]
  6. Bell, D.S. Type 2 diabetes mellitus: What is the optimal treatment regimen? Am. J. Med. 2004, 116, 23–29. [Google Scholar] [CrossRef]
  7. Rakesh, K.; Darshini, N.; Shubhavathi, T.; Mallesha, N. Biological Applications of Quinazolinone Analogues A Review. Org. Med. Chem. Int. J. 2017, 2, 41–45. [Google Scholar]
  8. Reddy, M.M.; Sivaramakrishna, A. Remarkably flexible quinazolinones—Synthesis and biological applications. J. Heterocycl. Chem. 2020, 57, 942–954. [Google Scholar] [CrossRef]
  9. Hussain, R.; Ullah, H.; Rahim, F.; Sarfraz, M.; Taha, M.; Iqbal, R.; Rehman, W.; Khan, S.; Shah, S.A.A.; Hyder, S. Multipotent Cholinesterase Inhibitors for the Treatment of Alzheimer’s Disease: Synthesis, Biological Analysis and Molecular Docking Study of Benzimidazole-Based Thiazole Derivatives. Molecules 2022, 27, 6087. [Google Scholar] [CrossRef]
  10. Parekh, N.M.; Juddhawala, K.V.; Rawal, B.M. Antimicrobial activity of thiazolylbenzenesulfonamide-condensed 2,4-thiazolidinediones derivatives. Med. Chem. Res. 2013, 22, 2737–2745. [Google Scholar] [CrossRef]
  11. Rostom, S.A.; El-Ashmawy, I.M.; Abd El Razik, H.A.; Badr, M.H.; Ashour, H.M. Design and synthesis of some thiazolyl and thiadiazolyl derivatives of antipyrine as potential non-acidic anti-inflammatory, analgesic and antimicrobial agents. Bioorg. Med. Chem. 2009, 17, 882–895. [Google Scholar] [CrossRef] [PubMed]
  12. Haroun, M.; Tratrat, C.; Tsolaki, E.; Geronikaki, A. Thiazole-based thiazolidinones as potent antimicrobial agents. Design, synthesis and biological evaluation. Comb. Chem. High Throughput Screen. 2016, 19, 51–57. [Google Scholar] [CrossRef]
  13. Mishra, I.; Mishra, R.; Mujwar, S.; Chandra, P.; Sachan, N. A retrospect on antimicrobial potential of thiazole scaffold. J. Heterocycl. Chem. 2020, 57, 2304–2329. [Google Scholar] [CrossRef]
  14. Luzina, E.L.; Popov, A.V. Synthesis and anticancer activity of N-bis (trifluoromethyl) alkyl-N′-thiazolyl and N-bis (trifluoromethyl) alkyl-N′-benzothiazolylureas. Eur. J. Med. Chem. 2009, 44, 4944–4953. [Google Scholar] [CrossRef] [PubMed]
  15. Zablotskaya, A.; Segal, I.; Germane, S.; Shestakova, I.; Domracheva, I.; Nesterova, A.; Geronikaki, A.; Lukevics, E. Silyl modification of biologically active compounds. 8. Trimethylsilyl ethers of hydroxyl-containing thiazole derivatives. Chem. Heterocycl. Compd. 2002, 38, 859–866. [Google Scholar] [CrossRef]
  16. Kalkhambkar, R.; Kulkarni, G.; Shivkumar, H.; Rao, R.N. Synthesis of novel triheterocyclicthiazoles as anti-inflammatory and analgesic agents. Eur. J. Med. Chem. 2007, 42, 1272–1276. [Google Scholar] [CrossRef] [PubMed]
  17. de Santana, T.I.; de Oliveira Barbosa, M.; de Moraes Gomes, P.A.T.; da Cruz, A.C.N.; da Silva, T.G.; Leite, A.C.L. Synthesis, anticancer activity and mechanism of action of new thiazole derivatives. Eur. J. Med. Chem. 2018, 144, 874–886. [Google Scholar] [CrossRef]
  18. Ayati, A.; Emami, S.; Moghimi, S.; Foroumadi, A. Thiazole in the targeted anticancer drug discovery. Future Med. Chem. 2019, 11, 1929–1952. [Google Scholar] [CrossRef]
  19. Britschgi, M.; Greyerz, S.; Burkhart, C.; Pichler, W.J. Molecular aspects of drug recognition by specific T cells. Curr. Drug Targets 2003, 4, 1–11. [Google Scholar] [CrossRef]
  20. Liaras, K.; Fesatidou, M.; Geronikaki, A. Thiazoles and thiazolidinones as COX/LOX inhibitors. Molecules 2018, 23, 685. [Google Scholar] [CrossRef]
  21. Turan-Zitouni, G.; Chevallet, P.; Kilic, F.S.; Erol, K. Synthesis of some thiazolyl-pyrazoline derivatives and preliminary investigation of their hypotensive activity. Eur. J. Med. Chem. 2000, 35, 635–641. [Google Scholar] [CrossRef] [PubMed]
  22. Haroon, M.; Khalid, M.; Shahzadi, K.; Akhtar, T.; Saba, S.; Rafique, J.; Ali, S.; Irfan, M.; Alam, M.M.; Imran, M. Alkyl 2-(2-(arylidene) alkylhydrazinyl) thiazole-4-carboxylates: Synthesis, acetyl cholinesterase inhibition and docking studies. J. Mol. Struct. 2021, 1245, 131063. [Google Scholar] [CrossRef]
  23. Chhabria, M.T.; Patel, S.; Modi, P.; Brahmkshatriya, P.S. Thiazole: A review on chemistry, synthesis and therapeutic importance of its derivatives. Curr. Top. Med. Chem. 2016, 16, 2841–2862. [Google Scholar] [CrossRef] [PubMed]
  24. Orlemans, E.; Verboom, W.; Scheltinga, M.; Reinhoudt, D.; Lelieveld, P.; Fiebig, H.; Winterhalter, B.; Double, J.; Bibby, M. Synthesis, mechanism of action, and biological evaluation of mitosenes. J. Med. Chem. 1989, 32, 1612–1620. [Google Scholar] [CrossRef]
  25. Belohlavek, D.; Malfertheiner, P. The effect of zolimidine, imidazopyridine-derivate, on the duodenal ulcer healing. Scand. J. Gastroenterol. Suppl. 1979, 54, 44. [Google Scholar]
  26. Gali, R.; Banothu, J.; Bavantula, R. One-Pot Multicomponent Synthesis of Novel Substituted Imidazo [1,2-a] Pyridine Incorporated Thiazolyl Coumarins and their Antimicrobial Activity. J. Heterocycl. Chem. 2015, 52, 641–646. [Google Scholar] [CrossRef]
  27. Althagafi, I.; Abdel-Latif, E. Synthesis and antibacterial activity of new imidazo [1,2-a] pyridines festooned with pyridine, thiazole or pyrazole moiety. Polycycl. Aromat. Compd. 2022, 42, 4487–4500. [Google Scholar] [CrossRef]
  28. Rahim, F.; Javed, M.T.; Ullah, H.; Wadood, A.; Taha, M.; Ashraf, M.; Khan, M.A.; Khan, F.; Mirza, S.; Khan, K.M. Synthesis, molecular docking, acetylcholinesterase and butyrylcholinesterase inhibitory potential of thiazole analogs as new inhibitors for Alzheimer disease. Bioorg. Chem. 2015, 62, 106–116. [Google Scholar] [CrossRef]
  29. Mekky, A.E.; Sanad, S.M.; El-Idreesy, T.T. New thiazole and thiazole-chromene hybrids possessing morpholine units: Piperazine-mediated one-pot synthesis of potential acetylcholinesterase inhibitors. Synth. Commun. 2021, 51, 3332–3344. [Google Scholar] [CrossRef]
  30. Hemaida, A.Y.; Hassan, G.S.; Maarouf, A.R.; Joubert, J.; El-Emam, A.A. Synthesis and biological evaluation of thiazole-based derivatives as potential acetylcholinesterase inhibitors. ACS Omega 2021, 6, 19202–19211. [Google Scholar] [CrossRef]
  31. Park, S.; Kim, H.; Son, J.-Y.; Um, K.; Lee, S.; Baek, Y.; Seo, B.; Lee, P.H. Synthesis of Imidazopyridines via Copper-Catalyzed, Formal Aza-[3 + 2] Cycloaddition Reaction of Pyridine Derivatives with α-Diazo Oxime Ethers. J. Org. Chem. 2017, 82, 10209–10218. [Google Scholar] [CrossRef] [PubMed]
  32. Mishra, N.P.; Mohapatra, S.; Sahoo, C.R.; Raiguru, B.P.; Nayak, S.; Jena, S.; Padhy, R.N. Design, one-pot synthesis, molecular docking study, and antibacterial evaluation of novel 2H-chromene based imidazo [1,2-a] pyridine derivatives as potent peptide deformylase inhibitors. J. Mol. Struct. 2021, 1246, 131183. [Google Scholar] [CrossRef]
  33. Thakur, A.; Pereira, G.; Patel, C.; Chauhan, V.; Dhaked, R.K.; Sharma, A. Design, one-pot green synthesis and antimicrobial evaluation of novel imidazopyridine bearing pyran bis-heterocycles. J. Mol. Struct. 2020, 1206, 127686. [Google Scholar] [CrossRef]
  34. Eskandani, M.; Babak Bahadori, M.; Zengin, G.; Dinparast, L.; Bahadori, S. Novel natural agents from Lamiaceae family: An evaluation on toxicity and enzyme inhibitory potential linked to diabetes mellitus. Curr. Bioact. Compd. 2016, 12, 34–38. [Google Scholar] [CrossRef]
  35. Khan, S.; Iqbal, S.; Rahim, F.; Shah, M.; Hussain, R.; Alrbyawi, H.; Rehman, W.; Dera, A.A.; Rasheed, L.; Somaily, H. New Biologically Hybrid Pharmacophore Thiazolidinone-Based Indole Derivatives: Synthesis, In Vitro Alpha-Amylase and Alpha-Glucosidase along with Molecular Docking Investigations. Molecules 2022, 27, 6564. [Google Scholar] [CrossRef]
  36. Khan, S.; Iqbal, S.; Shah, M.; Rehman, W.; Hussain, R.; Rasheed, L.; Alrbyawi, H.; Dera, A.A.; Alahmdi, M.I.; Pashameah, R.A. Synthesis, In Vitro Anti-Microbial Analysis and Molecular Docking Study of Aliphatic Hydrazide-Based Benzene Sulphonamide Derivatives as Potent Inhibitors of α-Glucosidase and Urease. Molecules 2022, 27, 7129. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Thiazole skeleton-containing biologically active drugs.
Figure 1. Thiazole skeleton-containing biologically active drugs.
Pharmaceuticals 16 01288 g001
Figure 2. Medicinal significance of drugs containing the imidazopyridine moiety.
Figure 2. Medicinal significance of drugs containing the imidazopyridine moiety.
Pharmaceuticals 16 01288 g002
Figure 3. Rationale of the current work.
Figure 3. Rationale of the current work.
Pharmaceuticals 16 01288 g003
Scheme 1. Synthesis of imidazopyridine-derived thiazole analogs.
Scheme 1. Synthesis of imidazopyridine-derived thiazole analogs.
Pharmaceuticals 16 01288 sch001
Figure 4. The protein–ligand interaction profile of the most potent scaffold 4g against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Figure 4. The protein–ligand interaction profile of the most potent scaffold 4g against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Pharmaceuticals 16 01288 g004
Figure 5. The PLI of the second-most active analog 4a against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Figure 5. The PLI of the second-most active analog 4a against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Pharmaceuticals 16 01288 g005
Figure 6. The PLI of the third-most active analog 4o against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Figure 6. The PLI of the third-most active analog 4o against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Pharmaceuticals 16 01288 g006
Figure 7. The PLI of thefourth-mostactive analog 4p against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Figure 7. The PLI of thefourth-mostactive analog 4p against the targeted α-glucosidase enzyme and its 3D and 2D diagram.
Pharmaceuticals 16 01288 g007
Figure 8. SAR studies of 4a, 4g, 4o, and 4p analogs.
Figure 8. SAR studies of 4a, 4g, 4o, and 4p analogs.
Pharmaceuticals 16 01288 g008
Figure 9. SAR studies of 4k, 4m, 4f, and 4e analogs.
Figure 9. SAR studies of 4k, 4m, 4f, and 4e analogs.
Pharmaceuticals 16 01288 g009
Figure 10. SAR studies of 4b, 4i, 4j, and 4l analogs.
Figure 10. SAR studies of 4b, 4i, 4j, and 4l analogs.
Pharmaceuticals 16 01288 g010
Figure 11. SAR studies of 4c, 4d, 4h, and 4n analogs.
Figure 11. SAR studies of 4c, 4d, 4h, and 4n analogs.
Pharmaceuticals 16 01288 g011
Graph 1. Representation of the drug-likeness properties of analog 4g.
Graph 1. Representation of the drug-likeness properties of analog 4g.
Pharmaceuticals 16 01288 g012
Graph 2. Representation of the drug-likeness properties of analog 4a.
Graph 2. Representation of the drug-likeness properties of analog 4a.
Pharmaceuticals 16 01288 g013
Graph 3. Representation of the drug-likeness properties of analog 4o.
Graph 3. Representation of the drug-likeness properties of analog 4o.
Pharmaceuticals 16 01288 g014
Graph 4. Representation of the drug-likeness properties of analog 4p.
Graph 4. Representation of the drug-likeness properties of analog 4p.
Pharmaceuticals 16 01288 g015
Table 1. Different substituent(s) and MIC (α-glucosidase) values of synthesized imidazopyridine incorporating thiazole scaffolds (4a-p).
Table 1. Different substituent(s) and MIC (α-glucosidase) values of synthesized imidazopyridine incorporating thiazole scaffolds (4a-p).
Pharmaceuticals 16 01288 i001
S.NORIC50 ± SEM a [μM]S.NORIC50 ± SEM a [μM]
4aPharmaceuticals 16 01288 i0026.85 ± 2.184iPharmaceuticals 16 01288 i00334.91 ± 5.84
4bPharmaceuticals 16 01288 i00413.63 ± 1.674jPharmaceuticals 16 01288 i00522.57 ± 3.55
4cPharmaceuticals 16 01288 i00663.46 ± 5.284kPharmaceuticals 16 01288 i00734.73 ± 5.23
4dPharmaceuticals 16 01288 i00832.12 ± 4.294lPharmaceuticals 16 01288 i00919.26 ± 2.58
4ePharmaceuticals 16 01288 i01044.89 ± 5.274mPharmaceuticals 16 01288 i01156.30 ± 5.94
4fPharmaceuticals 16 01288 i01250.96 ± 5.804nPharmaceuticals 16 01288 i01361.42 ± 4.56
4gPharmaceuticals 16 01288 i0145.57 ± 3.454oPharmaceuticals 16 01288 i0157.16 ± 1.40
4hPharmaceuticals 16 01288 i01643.20 ± 6.164pPharmaceuticals 16 01288 i01710.48 ± 2.20
a Standard Acarbose drug48.71 ± 2.65
Table 2. Representation of the types of interactions established by the most active analogs withthe active residue of the targeted enzyme from varied distances along with docking scores.
Table 2. Representation of the types of interactions established by the most active analogs withthe active residue of the targeted enzyme from varied distances along with docking scores.
Active AnalogsTargeted EnzymeReceptorsTypes of InteractionsDistance (oA)Docking Score
4gα-glucosidaseGLU-A-595Pi-Anion6.52−13.45
GLU-A-595Pi-Anion6.85
PHE-A-200Pi-Pi stacked5.69
PHE-A-200Pi-Pi stacked4.80
VAL-A-96H-B3.98
VAL-A-96Pi-R5.34
PHE-A-95Pi-R6.69
PHE-A-95Pi-R4.18
PHE-A-95H-F4.87
HIS-A-94Pi-R5.48
HIS-A-94H-F6.00
HIS-A-94H-F4.66
PHE-A-95C-H3.93
4aα-glucosidaseARG-A-91Pi-R5.47−12.87
TRP-A-90Pi-S7.20
TRP-A-90Pi-R6.58
PHE-A-200H-F4.04
PHE-A-200Pi-R5.41
VAL-A-201Pi-R4.94
VAL-A-96Pi-R4.88
HIS-A-94H-F5.80
HIS-A-94Pi-R5.84
GLN-A-202H-B4.09
4oα-glucosidaseGLU-A-595Pi-anion7.69−12.15
GLN-A-202H-B4.26
VAL-A-96Pi-R5.53
VAL-A-96Pi-R5.10
ARG-A-91Pi-R5.86
ARG-A-91Pi-R5.49
PHE-A-51Pi-Pi T-shaped5.47
HIS-A-94C-H6.18
HIS-A-94Pi-Pi T-shaped5.05
4pα-glucosidasePHE-A-51H-F6.98−11.25
HIS-A-94H-F4.45
HIS-A-94C-H3.79
GLU-A-595Pi-Anion5.99
GLU-A-595Pi-Anion6.83
VAL-A-201Pi-R6.06
PHE-A-200Pi-Pi T-shaped5.99
VAL-A-96Pi-R6.24
ARG-A-91Pi-R5.93
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hussain, R.; Rehman, W.; Khan, S.; Maalik, A.; Hefnawy, M.; Alanazi, A.S.; Khan, Y.; Rasheed, L. Imidazopyridine-Based Thiazole Derivatives as Potential Antidiabetic Agents: Synthesis, In Vitro Bioactivity, and In Silico Molecular Modeling Approach. Pharmaceuticals 2023, 16, 1288. https://doi.org/10.3390/ph16091288

AMA Style

Hussain R, Rehman W, Khan S, Maalik A, Hefnawy M, Alanazi AS, Khan Y, Rasheed L. Imidazopyridine-Based Thiazole Derivatives as Potential Antidiabetic Agents: Synthesis, In Vitro Bioactivity, and In Silico Molecular Modeling Approach. Pharmaceuticals. 2023; 16(9):1288. https://doi.org/10.3390/ph16091288

Chicago/Turabian Style

Hussain, Rafaqat, Wajid Rehman, Shoaib Khan, Aneela Maalik, Mohamed Hefnawy, Ashwag S. Alanazi, Yousaf Khan, and Liaqat Rasheed. 2023. "Imidazopyridine-Based Thiazole Derivatives as Potential Antidiabetic Agents: Synthesis, In Vitro Bioactivity, and In Silico Molecular Modeling Approach" Pharmaceuticals 16, no. 9: 1288. https://doi.org/10.3390/ph16091288

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop