Next Article in Journal
Identification of Iminium Intermediates Generation in the Metabolism of Tepotinib Using LC-MS/MS: In Silico and Practical Approaches to Bioactivation Pathway Elucidation
Previous Article in Journal
Distribution of Protein Precipitation Capacity within Variable Proanthocyanidin Fingerprints
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pyrazole Incorporated New Thiosemicarbazones: Design, Synthesis and Investigation of DPP-4 Inhibitory Effects

1
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
2
Medicinal Plant, Drug and Scientific Research and Application Center, Anadolu University, 26470 Eskişehir, Turkey
3
Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Anadolu University, 26470 Eskişehir, Turkey
*
Author to whom correspondence should be addressed.
Submission received: 15 October 2020 / Revised: 27 October 2020 / Accepted: 28 October 2020 / Published: 28 October 2020
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Dipeptidyl peptidase-4 (DPP-4) inhibition has been recognized as a promising approach to develop safe and potent antidiabetic agents for the management of type 2 diabetes. In this context, new thiosemicarbazones (2ao) were prepared efficiently by the reaction of aromatic aldehydes with 4-[4-(1H-pyrazol-1-yl)phenyl]thiosemicarbazide (1), which was obtained via the reaction of 4-(1H-pyrazol-1-yl)phenyl isothiocyanate with hydrazine hydrate. Compounds 2ao were evaluated for their DPP-4 inhibitory effects based on a convenient fluorescence-based assay. 4-[4-(1H-pyrazol-1-yl)phenyl]-1-(4-bromobenzylidene)thiosemicarbazide (2f) was identified as the most effective DPP-4 inhibitor in this series with an IC50 value of 1.266 ± 0.264 nM when compared with sitagliptin (IC50 = 4.380 ± 0.319 nM). MTT test was carried out to assess the cytotoxic effects of compounds 2ao on NIH/3T3 mouse embryonic fibroblast (normal) cell line. According to cytotoxicity assay, compound 2f showed cytotoxicity towards NIH/3T3 cell line with an IC50 value higher than 500 µM pointing out its favourable safety profile. Molecular docking studies indicated that compound 2f presented π-π interactions with Arg358 and Tyr666 via pyrazole scaffold and 4-bromophenyl substituent, respectively. Overall, in vitro and in silico studies put emphasis on that compound 2f attracts a great notice as a drug-like DPP-4 inhibitor for further antidiabetic research.

1. Introduction

Diabetes Mellitus (DM) is a complex, metabolic and chronic disorder and a serious threat to human health. DM is mainly divided into two types including type 1 or insulin-dependent DM (T1DM) and type 2 or non-insulin-dependent DM (T2DM). T2DM, which is generally attributed to insulin resistance in target cells and insufficient production of insulin from the β-cells, affects the majority of diabetic patients (around 90–95%) worldwide [1,2,3]. There are current therapy options for T2DM management such as sulphonylureas, glinides and insulin therapy, whereas these antidiabetic medications have an increased risk of hypoglycaemia, obesity and oedema. Therefore, there is an urgent need to discover new weight neutral and well-tolerated antidiabetic agents with a lower risk of hypoglycemia [4,5].
Incretin-based activity plays a pivotal role in the metabolic modulation of T2DM [6]. Glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide-1 (GLP-1) are incretin hormones secreted by intestinal endocrine cells in response to incoming nutrients and they are responsible for nearly 60% of the postprandial insulin release [7,8]. Based on the potent glucose-lowering actions of these hormones, GLP-1 receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors have been developed as incretin mimetics and incretin enhancers, respectively [9,10].
DPP-4 is a serine exopeptidase, which is able to cleave dipeptides from polypeptides such as incretin hormones, some chemokines and cytokines with a proline or alanine amino acid in the penultimate position at the N-terminus of the peptide chain. This outcome indicates that DPP-4 is not only implicated in glucose homeostasis but also in neurogenic inflammation, blood pressure and the immune system [8,11,12,13,14]. DPP-4 inhibitors have been reported to stimulate insulin secretion and suppress glucagon secretion in a glucose-dependent way causing a low risk of hypoglycaemia and weight gain compared to other antidiabetic agents. “Gliptins” such as sitagliptin, linagliptin and vildagliptin are well-known DPP-4 inhibitors (Figure 1). Among them, sitagliptin and vildagliptin improve glycaemic control and reduce glycosylated haemoglobin (HbA1c) level in monotherapy or combined therapy with metformin and thiazolidinediones. Some gliptins also possess protective properties in kidney functions and cardiovascular system [15,16,17,18].
Thiosemicarbazones have gained extensive interest, in particular with their capacity of forming metal ion coordination modes and metal complexes. They are one of the most important members of N,S donor ligands and have been reported to show a wide pharmacological utility including antidiabetic activity. There are also studies suggesting thiosemicarbazones as aldose reductase and glycogen phosphorylase inhibitors to be effective in T2DM treatment [19,20,21,22,23,24]. Among these studies, Alexacou et al. synthesized some new β-d-glucopyranosyl-thiosemicarbazone derivatives and investigated their glycogen phosphorylase inhibitory effects by kinetic experiments and crystallographic binding studies. Para fluorophenyl substituted compound was identified as the most potent glycogen phosphorylase inhibitor (IC50 = 5.7 µM) (Figure 2). In another study, Kulkarni et al. prepared new transition metal complexes of quinoxaline–thiosemicarbazone ligands and investigated their antidiabetic activity using blood-glucose and oral glucose tolerance tests. They found that some of them showed significant antidiabetic effects with low toxicity and a high safety profile (Figure 2). On the other hand, Shehzad et al. reported a new series of benzoxazinone-thiosemicarbazones as aldose reductase inhibitors. Meta floro and bromophenyl substituted compounds revealed the most significant aldose reductase inhibition (Figure 2) [20,21,24].
Pyrazole is a five-membered, heterocyclic compound with two adjacent nitrogen atoms. Pyrazole nucleus can react with both acids and bases due to its distinct nitrogen atoms exhibiting “pyrrole-like” (N1) and “pyridine-like” (N2) behaviours. Pyrazole derivatives have been reported to exhibit a broad spectrum of pharmacological properties including antidiabetic activity and it is also found in a large variety of biologically active molecules. They are prominent competitive inhibitors or activators for antidiabetic drug design such as sodium-glucose co-transporter-1, sodium-glucose cotransporter-2, α-glucosidase, α-amylase and DPP-4 inhibitors, glucagon receptor antagonists and peroxisome proliferator-activated agonists [25,26,27,28,29,30,31,32]. Among these distinct types of antidiabetic activity, there are studies reporting pyrazole-based compounds as DPP-4 inhibitors. The extensive efforts of Yoshida et al. led to the discovery of tenegliptin, which has been approved for the treatment of type 2 diabetes in Japan (Figure 3). Besides, Hsu et al. also designed (1,3-diphenyl-1H-pyrazol-4-yl)methylamine analogues as DPP-4 inhibitors, and meta fluoro substituted compound was found to slightly increase DPP-4 inhibition according to in vitro studies (Figure 3). In another study, Wu et al. determined a lead compound after bioassay verification and they synthesized analogues of this lead compound. They showed that nine compounds exhibited DPP-4 inhibition. Naphthalen-1-yl bearing compound displayed the most potent inhibitory effect (Figure 3) [33,34,35].
Prompted by the aforementioned findings, pyrazole-based new thiosemicarbazones (2ao) were designed and synthesized. The DPP-4 inhibitory effects of all these compounds were determined and they were further evaluated for their cytotoxicities. Moreover, in silico molecular docking and pharmacokinetic studies were carried out to shed light on their mechanistic effects and pharmacokinetic profiles, respectively.

2. Results and Discussion

2.1. Chemistry

The synthesis of a new series of thiosemicarbazones (2ao) was performed as depicted in Scheme 1. The final compounds were obtained by the treatment of aromatic aldehydes with 4-[4-(1H-pyrazol-1-yl)phenyl]thiosemicarbazide (1), which was synthesized via the reaction of 4-(1H-pyrazol-1-yl)phenyl isothiocyanate with hydrazine hydrate.
The structures of compounds 2ao were confirmed by different spectroscopic techniques, namely Infrared (IR), 1H Nuclear Magnetic Resonance (NMR), 13C NMR, and High-Resolution Mass Spectrometry (HRMS).
In the IR spectra of compounds 2ao, the characteristic N-H stretching vibrations belonging to the thiosemicarbazone moiety were observed in the region 3346.50–3282.84 cm−1.
In the 1H NMR spectra of compounds 2ao, both NH protons of thiosemicarbazone moiety were determined as singlet peaks at 10.03–10.36 ppm and 11.69–12.11 ppm. The characteristic azomethine (-CH=N-) protons appeared as a singlet at 8.22–8.51 ppm indicating, the formation of the thiosemicarbazone moiety of the final compounds. In their 13C NMR spectra, the characteristic carbons at 175.61–176.52 ppm and 140.80–144.65 ppm belonging to thiocarbonyl and azomethine groups, respectively, confirmed the formation of the thiosemicarbazone moiety of the final compounds. Moreover, the C3, C4 and C5 carbons of the pyrazole scaffold of the final compounds gave rise to the peaks at 140.40–141.36 ppm, 107.67–108.28 ppm and 121.60–131.24 ppm, respectively (Figure 4). Finally, the HRMS data of all compounds coincided with their molecular formulas.

2.2. Biological Activity

All synthesized compounds (2ao) were screened for their in vitro DPP-4 inhibitory potencies at 100 µM concentration using a fluorescence-based assay (Table 1). Non-substituted compound 2a caused the lowest DPP-4 inhibition (20.67 ± 1.82%), whilst bromo-substituted compound 2f showed DPP-4 inhibition (98.20 ± 0.89%) even more than sitagliptin. Compounds 2f, 2g, 2i, 2k, and 2o, which exhibited DPP-4 inhibition above 60%, were further analyzed for the determination of their half-maximal inhibitory concentration (IC50) values (Table 2).
Bromo-substituted compound 2f was the most potent DPP-4 inhibitor in this series with an IC50 value of 1.266 ± 0.264 nM as indicated in Table 2. This compound was found to be more active than sitagliptin (IC50 = 4.380 ± 0.319 nM) indicating that the introduction of the bromo substituent into the para position of the benzylidene moiety gave rise to a substantial increase in DPP-4 inhibitory potency. The replacement of the bromo substituent of compound 2f with the fluoro or the chloro substituent led to a significant drop in DPP-4 inhibitory activity.
Trifluoromethyl-substituted compound 2g showed marked inhibitory effect on DPP-4 with an IC50 value of 4.775 ± 0.296 nM, similar to sitagliptin. It can be concluded that its DPP-4 inhibitory potency results from the presence of the trifluoromethyl group at the para position of the benzylidene moiety. Biphenyl-substituted compound 2o and thiomethyl-substituted compound 2k displayed moderate inhibitory effects on DPP-4 with IC50 values of 18.061 ± 0.311 nM and 22.671 ± 0.301 nM, respectively. Compound 2i bearing a methoxy group showed DPP-4 inhibitory potency with an IC50 value of 43.312 ± 0.372 nM.
In an attempt to assess whether the compounds were toxic or non-toxic to normal cells, compounds 2ao were investigated for their cytotoxic effects on NIH/3T3 mouse embryonic fibroblast (normal) cells by means of MTT assay. All derivatives showed cytotoxicity towards the NIH/3T3 cell line with IC50 values higher than 500 µM. It can be concluded that they possess favourable safety profiles.

2.3. In Silico Studies

Molecular docking studies were conducted for compounds 2f, 2g, 2i, 2k, and 2o in the active site of DPP-4 (PDB ID: 4FFW) compared to sitagliptin as they were defined as the most potent DPP-4 inhibitors based on the in vitro enzyme studies. The results of molecular docking indicated that substitutions on the thiosemicarbazone framework directly affected the proper interactions. In general, all these compounds formed cation-π interactions with Arg358 and Tyr666, which are key components in specific S1 and S2 pockets of DDP-4 [8] through pyrazole moiety and substituents at the para position of the phenyl ring (Figure 5). The bromo and the trifluoromethyl substitutions played major roles in the high activity of compounds 2f and 2g associated with their strong π-π interactions with Arg358 (Figure 6). Sitagliptin also presented the same strong interactions with Arg358 and Tyr666, whereas it also revealed hydrogen bonding and salt bridge formation with Glu205 and Glu206, which hampers its high DPP-4 inhibition. Compounds 2i and 2k, which were found less active DPP-4 inhibitors related to in vitro studies compared to compounds 2f, 2g, and 2o, also formed hydrogen bonding with Glu205. These results were found totally coherent with in vitro studies.
Some Absorption, Distribution, Metabolism, and Excretion (ADME) properties including predicted octanol/water partition coefficient (QPlogPo/w), the conformation-independent predicted aqueous solubility (CIQPlogS), brain/blood partition coefficient (QPlogBB), human serum albumin binding (QPlogKhsa), and predicted human oral absorption were investigated in silico. The results given in Table 3 were found within the acceptable range based on the specified parameters including their predicted human oral absorption rates making these compounds appropriate drug candidates. Besides, compounds 2ao had no more than one violation of the certain criteria of Lipinski’s rule of five and Jorgensen’s rule of three.

3. Materials and Methods

3.1. Chemistry

All reagents purchased from commercial suppliers were used without further purification. The Electrothermal IA9200 digital melting point apparatus (Staffordshire, UK) was used to determine the melting points (M.p.) of the compounds. IR spectra were recorded on an IRPrestige-21 Fourier Transform Infrared spectrophotometer (Shimadzu, Tokyo, Japan). 1H NMR and 13C NMR spectra were recorded on Bruker 400 MHz and 500 MHz spectrometers (Bruker, Billerica, MA, USA). HRMS spectra were recorded on a Shimadzu LCMS-IT-TOF system (Shimadzu, Kyoto, Japan).

3.1.1. General Procedure for the Synthesis of the Compounds

[4-(1H-Pyrazol-1-yl)phenyl]thiosemicarbazide (1)

A mixture of 4-(1H-pyrazol-1-yl)phenyl isothiocyanate (0.1 mol) and hydrazine hydrate (0.2 mol) in ethanol (30 mL) was stirred at room temperature for 4 h and then filtered. The residue was crystallized from ethanol [36].

[4-(1H-Pyrazol-1-yl)phenyl]-1-(arylidene)thiosemicarbazide (2ao)

A mixture of 4-[4-(1H-pyrazol-1-yl)phenyl]thiosemicarbazide (1) (0.01 mol) and aromatic aldehyde (0.01 mol) was refluxed in ethanol for 8 h, filtered and crystallized from ethanol [36].

[4-(1H-Pyrazol-1-yl)phenyl]-1-(benzylidene)thiosemicarbazide (2a)

Off-white powder. M.p.: 218–220 °C. Yield: 75%.
IR νmax (cm−1): 3298.28 (N-H stretching), 3149.76, 3116.97 (Aromatic C-H stretching), 2991.59, 2941.44 (Aliphatic C-H stretching), 1597.06, 1541.12, 1523.76, 1502.55 (N-H bending, C=N and C=C stretching), 1392.61, 1336.67, 1269.16, 1199.72, 1122.57, 1053.13, 1029.99 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 993.55, 829.39, 771.53, 758.02, 725.23, 688.59 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S1).
1H NMR (400 MHz, DMSO-d6): 6.54 (s, 1H), 7.43–7.45 (m, 3H), 7.74 (d, J = 8.80 Hz, 3H), 7.85 (d, J = 8.40 Hz, 2H), 7.91–7.93 (m, 2H), 8.22 (s, 1H), 8.47 (s, 1H), 10.18 (s, 1H), 11.89 (s, 1H) (Supplementary Material Figures S2–S4).
13C NMR (100 MHz, DMSO-d6): 108.28 (CH), 117.96 (2CH), 126.60 (2CH), 127.54 (CH), 127.56 (2CH), 128.54 (2CH), 129.98 (CH), 133.91 (C), 136.84 (C), 137.06 (C), 140.76 (CH), 143.04 (CH), 176.04 (C) (Supplementary Material Figure S5).
HRMS (ESI) (m/z) [M + H]+ calcd. for C17H15N5S: 322.1121, found: 322.1122 (Supplementary Material Figure S6).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-nitrobenzylidene)thiosemicarbazide (2b)

Yellow powder. M.p.: 244–246 °C. Yield: 90%.
IR νmax (cm−1): 3313.71 (N-H stretching), 3149.76, 3099.61, 3049.46 (Aromatic C-H stretching), 2916.37, 2835.36 (Aliphatic C-H stretching), 1581.63, 1537.27, 1510.26, 1483.26 (N-H bending, C=N and C=C stretching), 1433.11, 1409.96, 1394.53, 1330.88, 1284.59, 1253.73, 1168.86, 1101.35, 1031.92 (C-H bending, NO2, C=S and C-N stretching, aromatic C-H in plane bending), 935.48, 873.75, 831.32, 746.45, 688.59, 659.66 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S7).
1H NMR (400 MHz, DMSO-d6): 6.52 (s, 1H), 7.68 (d, J = 8.40 Hz, 2H), 7.73 (s, 1H), 7.83 (d, J = 8.40 Hz, 2H), 8.17 (d, J = 8.40 Hz, 3H), 8.23 (d, J = 8.40 Hz, 2H), 8.46 (s, 1H), 10.33 (s, 1H), 12.10 (s, 1H) (Supplementary Material Figures S8–S10).
13C NMR (100 MHz, DMSO-d6): 107.70 (CH), 117.99 (2CH), 123.64 (2CH), 126.89 (2CH), 127.57 (CH), 128.39 (2CH), 136.88 (C), 137.05 (C), 140.27 (C), 140.40 (CH), 140.80 (CH), 147.67 (C), 176.52 (C, C=S) (Supplementary Material Figure S11).
HRMS (ESI) (m/z) [M + H]+ calcd. for C17H14N6O2S: 367.0972, found: 367.0984) (Supplementary Material Figure S12).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-cyanobenzylidene)thiosemicarbazide (2c)

Green powder. M.p.: 226–229 °C. Yield: 88%.
IR νmax (cm−1): 3313.07 (N-H stretching), 3136.25 (Aromatic C-H stretching), 2981.95, 2848.86 (Aliphatic C-H stretching), 2218.14 (C≡N stretching), 1622.13, 1598.99, 1537.27, 1525.69, 1500.62 (N-H bending, C=N and C=C stretching), 1435.04, 1394.53, 1332.81, 1276.88, 1207.44, 1168.86, 1082.07, 1041.56, 1029.99 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 933.55, 867.97, 831.32, 777.31, 750.31, 671.23 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S13).
1H NMR (400 MHz, DMSO-d6): 6.55 (s, 1H), 7.67 (d, J = 8.80 Hz, 3H), 7.75–7.90 (m, 4H), 8.14 (d, J = 8.80 Hz, 2H), 8.20 (s, 1H), 8.51 (s, 1H), 10.35 (s, 1H), 12.10 (s, 1H) (Supplementary Material Figures S14–S16).
13C NMR (100 MHz, DMSO-d6): 107.79 (CH), 111.70 (C), 117.99 (2CH), 118.74 (C), 127.05 (2CH), 127.66 (CH), 128.14 (2CH), 132.46 (2CH), 136.92 (C), 137.06 (C), 138.55 (C), 140.80 (CH), 140.88 (CH), 176.46 (C) (Supplementary Material Figure S17).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H14N6S: 347.1073, found: 347.1076 (Supplementary Material Figure S18).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-fluorobenzylidene)thiosemicarbazide (2d)

Beige powder. M.p.: 200–204 °C. Yield: 80%.
IR νmax (cm−1): 3282.84 (N-H stretching), 3142.04 (Aromatic C-H stretching), 2983.88, 2918.30 (Aliphatic C-H stretching), 1600.92, 1543.05, 1523.76, 1504.48 (N-H bending, C=N and C=C stretching), 1415.75, 1392.61, 1317.38, 1253.73, 1230.58, 1188.15, 1155.36, 1118.71, 1070.49, 1049.28, 1012.63 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 935.48, 871.82, 829.39, 796.60 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S19).
1H NMR (400 MHz, DMSO-d6): 6.53 (s, 1H), 7.27 (t, J = 8.40 Hz, 8.80 Hz, 17.20 Hz, 2H), 7.73 (t, J = 8.80 Hz, 17.60 Hz, 3H), 7.85 (d, J = 8.80 Hz, 2H), 7.94–8.02 (m, 2H), 8.19 (s, 1H), 8.48 (s, 1H), 10.23 (s, 1H), 11.93 (s, 1H) (Supplementary Material Figures S20–S22).
13C NMR (100 MHz, DMSO-d6): 107.77 (CH), 115.66 (2CH, d, J = 21.80 Hz), 118.01 (2CH), 126.84 (2CH), 127.61 (CH), 129.87 (2CH, d, J = 8.40 Hz), 130.60 (C), 136.93 (C), 137.10 (C), 140.85 (CH), 141.89 (CH), 161.93 (C), 176.08 (C) (Supplementary Material Figure S23).
HRMS (ESI) (m/z) [M + H]+ calcd. for C17H14FN5S: 340.1027, found: 340.1031 (Supplementary Material Figure S24).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-chlorobenzylidene)thiosemicarbazide (2e)

Off-white powder. M.p.: 194–198 °C. Yield: 86%.
IR νmax (cm−1): 3313.71 (N-H stretching), 3180.62, 3107.32, 3064.89, 3047.53 (Aromatic C-H stretching), 2995.45, 2941.44, 2920.23, 2848.86 (Aliphatic C-H stretching), 1593.20, 1523.76, 1481.33 (N-H bending, C=N and C=C stretching), 1433.11, 1400.32, 1390.68, 1315.45, 1280.73, 1255.66, 1182.36, 1168.86, 1082.07, 1041.56, 1010.70 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 931.62, 835.18, 813.96, 767.67, 732.95 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S25).
1H NMR (400 MHz, DMSO-d6): 6.54 (s, 1H), 7.49 (d, J = 8.40 Hz, 2H), 7.72 (t, J = 9.20 Hz, 9.60 Hz, 18.80 Hz, 3H), 7.84 (d, J = 8.40 Hz, 2H), 7.96 (d, J = 8.40 Hz, 2H), 8.18 (s, 1H), 8.48 (s, 1H), 10.22 (s, 1H), 11.92 (s, 1H) (Supplementary Material Figures S26–S28).
13C NMR (100 MHz, DMSO-d6): 107.67 (CH), 117.95 (2CH), 126.71 (2CH), 127.54 (CH), 128.60 (2CH), 129.20 (2CH), 132.90 (C), 134.45 (C), 136.89 (C), 137.01 (C), 140.76 (CH), 141.61 (CH), 176.13 (C) (Supplementary Material Figure S29).
HRMS (ESI) (m/z) [M + H]+ calcd. for C17H14ClN5S: 356.0731, found: 356.0740 (Supplementary Material Figure S30).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-bromobenzylidene)thiosemicarbazide (2f)

Beige powder. M.p.: 196–199 °C. Yield: 83%.
IR νmax (cm−1): 3315.63, 3288.63 (N-H stretching), 3140.11 (Aromatic C-H stretching), 2987.74, 2954.95, 2916.37, 2848.86 (Aliphatic C-H stretching), 1600.92, 1587.42, 1537.27, 1479.40, 1462.04 (N-H bending, C=N and C=C stretching), 1396.46, 1317.38, 1286.52, 1253.73, 1182.36, 1107.14, 1064.71, 1043.49, 1006.84 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 929.69, 835.18, 817.82, 767.67, 732.95 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S31).
1H NMR (400 MHz, DMSO-d6): 6.55 (s, 1H), 7.57–7.78 (m, 5H), 7.81–7.90 (m, 4H), 8.07 and 8.14 (2s, 1H), 8.50 (s, 1H), 10.24 (s, 1H), 11.94 (s, 1H) (Supplementary Material Figures S32–S35).
13C NMR (100 MHz, DMSO-d6): 107.77 (CH), 117.96 (2CH), 123.30 (C), 126.89 (2CH), 127.64 (CH), 129.38 (CH), 129.51 (CH), 131.58 (CH), 131.61 (CH), 131.99 (C), 136.94 (C), 137.03 (C), 140.85 (CH), 141.73 (CH), 176.15 (C) (Supplementary Material Figure S36).
HRMS (ESI) (m/z) [M + H]+ calcd. for C17H14BrN5S: 400.0226, found: 400.0238 (Supplementary Material Figure S37).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-trifluoromethylbenzylidene)thiosemicarbazide (2g)

White powder. M.p.: 222–225 °C. Yield: 79%.
IR νmax (cm−1): 3315.63 (N-H stretching), 3182.55, 3113.11, 3082.25 (Aromatic C-H stretching), 2918.30, 2848.86 (Aliphatic C-H stretching), 1602.85, 1529.55, 1487.12, 1454.33 (N-H bending, C=N and C=C stretching), 1433.11, 1392.61, 1323.17, 1284.59, 1259.52, 1180.44, 1155.36, 1095.57, 1064.71, 1029.99, 1012.63 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 933.55, 831.32, 804.32, 736.81 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S38).
1H NMR (400 MHz, DMSO-d6): 6.55 (s, 1H), 7.70 (d, J = 8.80 Hz, 2H), 7.75–7.79 (m, 3H), 7.86 (d, J = 8.80 Hz, 2H), 8.16 (d, J = 8.00 Hz, 2H), 8.25 (s, 1H), 8.50 (s, 1H), 10.34 (s, 1H), 12.09 (s, 1H) (Supplementary Material Figures S39–S41).
13C NMR (100 MHz, DMSO-d6): 107.77 (CH), 118.01 (2CH), 125.41 (C), 127.02 (2CH), 127.64 (2CH), 128.16 (2CH), 129.43 (CH), 129.74 (C), 136.99 (C), 137.06 (C), 138.04 (C), 140.87 (CH), 141.15 (CH), 176.46 (C) (Supplementary Material Figure S42).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H14F3N5S: 390.0995, found: 390.0997 (Supplementary Material Figure S43).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-methylbenzylidene)thiosemicarbazide (2h)

White powder. M.p.: 192–194 °C. Yield: 76%.
IR νmax (cm−1): 3302.13 (N-H stretching), 3157.47, 3026.31 (Aromatic C-H stretching), 2991.59, 2916.37 (Aliphatic C-H stretching), 1598.99, 1556.55, 1519.91, 1504.48 (N-H bending, C=N and C=C stretching), 1433.11, 1400.32, 1325.10, 1271.09, 1192.01, 1118.71, 1070.49, 1028.06 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 933.55, 871.82, 829.39, 808.17, 736.81, 686.66 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S44).
1H NMR (400 MHz, DMSO-d6): 2.34 (s, 3H), 6.55 (s, 1H), 7.25 (d, J = 8.00 Hz, 2H), 7.72–7.85 (m, 7H), 8.16 (s, 1H), 8.50 (s, 1H), 10.16 (s, 1H), 11.85 (s, 1H) (Supplementary Material Figures S45–S47).
13C NMR (100 MHz, DMSO-d6): 21.06 (CH3), 107.75 (CH), 117.94 (2CH), 126.73 (2CH), 127.63 (2CH), 129.25 (2CH), 131.24 (CH), 136.84 (C), 137.12 (C), 139.92 (2C), 140.83 (CH), 143.18 (CH), 175.86 (C) (Supplementary Material Figure S48).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H17N5S: 336.1277, found: 336.1282 (Supplementary Material Figure S49).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-methoxybenzylidene)thiosemicarbazide (2i)

Beige powder. M.p.: 190–192 °C. Yield: 78%.
IR νmax (cm−1): 3304.06 (N-H stretching), 3182.55, 3124.68 (Aromatic C-H stretching), 2999.31, 2918.30, 2848.86 (Aliphatic C-H stretching), 1606.70, 1597.06, 1543.05, 1523.76, 1504.48, 1481.33 (N-H bending, C=N and C=C stretching), 1388.75, 1334.74, 1303.88, 1253.73, 1184.29, 1168.86, 1120.64, 1093.64, 1024.20 (C-H bending, C=S, C-O and C-N stretching, aromatic C-H in plane bending), 931.62, 833.25, 786.96, 758.02, 665.44 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S50).
1H NMR (400 MHz, DMSO-d6): 3.81 (s, 3H), 6.54 (s, 1H), 6.99 (d, J = 8.40 Hz, 2H), 7.67–7.88 (m, 7H), 8.14 (s, 1H), 8.48 (s, 1H), 10.12 (s, 1H), 11.79 (s, 1H) (Supplementary Material Figures S51–S53).
13C NMR (100 MHz, DMSO-d6): 58.60 (CH3), 107.74 (CH), 114.13 (CH), 117.94 (2CH), 126.51 (CH), 126.64 (2CH), 127.57 (CH), 127.63 (CH), 129.12 (CH), 129.32 (C), 136.79 (C), 137.17 (C), 140.82 (CH), 143.04 (CH), 160.88 (C), 175.61 (C) (Supplementary Material Figure S54).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H17N5OS: 352.1227, found: 352.1229 (Supplementary Material Figure S55).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-methylsulfonylbenzylidene)thiosemicarbazide (2j)

Buff powder. M.p.: 230–234 °C. Yield: 82%.
IR νmax (cm−1): 3344.57 (N-H stretching), 3149.76, 3124.68 (Aromatic C-H stretching), 2995.45, 2916.37, 2848.86 (Aliphatic C-H stretching), 1598.99, 1525.69, 1502.55 (N-H bending, C=N and C=C stretching), 1433.11, 1390.68, 1332.81, 1303.88, 1288.45, 1269.16, 1193.94, 1138.00, 1080.14, 1043.49 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 968.27, 931.62, 869.90, 833.25, 823.60, 779.24 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S56).
1H NMR (400 MHz, DMSO-d6): 3.27 (s, 3H), 6.55 (s, 1H), 7.69 (d, J = 9.20 Hz, 2H), 7.76 (s, 1H), 7.86 (d, J = 8.80 Hz, 2H), 7.97 (d, J = 8.80 Hz, 2H), 8.20 (d, J = 8.40 Hz, 2H), 8.25 (s, 1H), 8.50 (s, 1H), 10.36 (s, 1H), 12.11 (s, 1H) (Supplementary Material Figures S57–S59).
13C NMR (100 MHz, DMSO-d6): 43.42 (CH3), 107.82 (CH), 118.03 (2CH), 127.11 (2CH), 127.20 (CH), 127.66 (2CH), 128.20 (2CH), 129.16 (C), 136.95 (C), 137.10 (C), 138.90 (C), 140.90 (CH), 141.24 (CH), 176.50 (C) (Supplementary Material Figure S60).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H17N5O2S2: 400.0896, found: 400.0902 (Supplementary Material Figure S61).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-methylthiobenzylidene)thiosemicarbazide (2k)

Pale yellow powder. M.p.: 196–198 °C. Yield: 84%.
IR νmax (cm−1): 3346.50 (N-H stretching), 3140.11, 3041.74 (Aromatic C-H stretching), 2983.88, 2912.51 (Aliphatic C-H stretching), 1595.13, 1541.12, 1523.76, 1490.97 (N-H bending, C=N and C=C stretching), 1435.04, 1400.32, 1332.81, 1273.02, 1197.79, 1120.64, 1087.85, 1039.63 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 933.55, 866.04, 831.32, 804.32, 769.60, 723.31 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S62).
1H NMR (500 MHz, DMSO-d6): 2.49 (s, 3H), 6.53 (s, 1H), 7.28 (d, J = 8.40 Hz, 2H), 7.69–7.74 (m, 3H), 7.82-7.85 (m, 4H), 8.13 (s, 1H), 8.48 (s, 1H), 10.16 (s, 1H), 11.85 (s, 1H) (Supplementary Material Figures S63–S65).
13C NMR (125 MHz, DMSO-d6): 14.74 (CH3), 108.27 (CH), 118.27 (2CH), 125.90 (CH), 127.27 (2CH), 128.14 (2CH), 128.56 (2CH), 130.89 (C), 137.35 (C), 137.61 (C), 141.34 (CH), 141.50 (CH), 143.17 (C), 176.30 (C) (Supplementary Material Figure S66).
HRMS (ESI) (m/z) [M + H]+ calcd. for C18H17N5S2: 368.0998, found: 368.1003 (Supplementary Material Figure S67).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-dimethylaminobenzylidene)thiosemicarbazide (2l)

Pale brown powder. M.p.: 210–213 °C. Yield: 85%.
IR νmax (cm−1): 3115.04, 3059.10 (Aromatic C-H stretching), 2974.23, 2895.15, 2806.43 (Aliphatic C-H stretching), 1606.70, 1543.05, 1521.84, 1510.26, 1444.68 (N-H bending, C=N and C=C stretching), 1394.53, 1357.89, 1330.88, 1267.23, 1215.15, 1170.79, 1118.71, 1070.49, 1049.28, 1033.85 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 939.33, 846.75, 819.75, 750.31, 734.88 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S68).
1H NMR (500 MHz, DMSO-d6): 2.97 (s, 6H), 6.55 (s, 1H), 6.73 (d, J = 7.08 Hz, 2H), 7.72 (d, J = 7.00 Hz, 2H), 7.75 (d, J = 7.08 Hz, 2H), 7.84 (d, J = 7.08 Hz, 3H), 8.08 (s, 1H), 8.50 (s, 1H), 10.03 (s, 1H), 11.69 (s, 1H) (Supplementary Material Figures S69–72).
13C NMR (125 MHz, DMSO-d6): 40.33 (2CH3), 108.23 (CH), 112.09 (2CH), 118.43 (2CH), 121.60 (CH), 126.86 (2CH), 128.10 (2CH), 129.56 (C), 137.13 (C), 137.77 (C), 141.30 (CH), 144.65 (CH), 152.02 (C), 175.39 (C) (Supplementary Material Figure S73).
HRMS (ESI) (m/z) [M + H]+ calcd. for C19H20N6S: 365.1543, found: 365.1547 (Supplementary Material Figure S74).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-isopropylbenzylidene)thiosemicarbazide (2m)

White powder. M.p.: 208–211 °C. Yield: 74%.
IR νmax (cm−1): 3115.04, 3064.89 (Aromatic C-H stretching), 2970.38, 2954.95, 2868.15 (Aliphatic C-H stretching), 1606.70, 1525.69, 1504.48, 1462.04 (N-H bending, C=N and C=C stretching), 1394.53, 1332.81, 1265.30, 1209.37, 1197.79, 1078.21, 1049.28, 1012.63 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 937.40, 846.75, 833.25, 775.38, 754.17, 732.95, 659.66 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S75).
1H NMR (500 MHz, DMSO-d6): 1.22 (d, J = 5.52 Hz, 6H), 2.89–2.95 (m, 1H), 6.56 (s, 1H), 7.31 (d, J = 6.56 Hz, 2H), 7.72 (d, J = 7.08 Hz, 2H), 7.76 (s, 1H), 7.83–7.86 (m, 4H), 8.17 (s, 1H), 8.51 (s, 1H), 10.17 (s, 1H), 11.88 (s, 1H) (Supplementary Material Figures S76–S79).
13C NMR (125 MHz, DMSO-d6): 24.14 (2CH3), 33.88 (CH), 108.26 (CH), 118.44 (2CH), 127.09 (CH), 127.21 (2CH), 128.13 (2CH), 128.25 (2CH), 132.15 (C), 137.34 (C), 137.60 (C), 141.33 (CH), 143.64 (CH), 151.23 (C), 176.35 (C) (Supplementary Material Figure S80).
HRMS (ESI) (m/z) [M + H]+ calcd. for C20H21N5S: 364.1590, found: 364.1593 (Supplementary Material Figure S81).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-tert-butylbenzylidene)thiosemicarbazide (2n)

White powder. M.p.: 235–238 °C. Yield: 72%.
IR νmax (cm−1): 3120.82 (Aromatic C-H stretching), 2960.73, 2954.95, 2866.22 (Aliphatic C-H stretching), 1606.70, 1537.27, 1525.69, 1506.41 (N-H bending, C=N and C=C stretching), 1394.53, 1334.74, 1265.30, 1213.23, 1122.57, 1080.14, 1049.28 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 937.40, 835.18, 759.95, 729.09, 661.58, 617.22 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S82).
1H NMR (500 MHz, DMSO-d6): 1.31 (s, 9H), 6.56 (s, 1H), 7.46 (d, J = 6.68 Hz, 2H), 7.72 (d, J = 7.08 Hz, 2H), 7.76 (s, 1H), 7.84 (d, J = 6.94 Hz, 4H), 8.17 (s, 1H), 8.51 (s, 1H), 10.15 (s, 1H), 11.87 (s, 1H) (Supplementary Material Figures S83–S86).
13C NMR (125 MHz, DMSO-d6): 31.43 (3CH3), 35.08 (C), 108.26 (CH), 118.45 (2CH), 125.91 (CH), 127.15 (2CH), 127.97 (2CH), 128.14 (2CH), 131.76 (C), 137.33 (C), 137.60 (C), 141.33 (CH), 143.54 (CH), 153.40 (C), 176.34 (C) (Supplementary Material Figure S87).
HRMS (ESI) (m/z) [M + H]+ calcd. for C21H23N5S: 378.1747, found: 378.1749 (Supplementary Material Figure S88).

[4-(1H-Pyrazol-1-yl)phenyl]-1-(4-phenylbenzylidene)thiosemicarbazide (2o)

Yellow powder. M.p.: 210–212 °C. Yield: 83%.
IR νmax (cm−1): 3311.78 (N-H stretching), 3132.40, 3030.17 (Aromatic C-H stretching), 2978.09 (Aliphatic C-H stretching), 1597.06, 1537.27, 1523.76, 1496.76 (N-H bending, C=N and C=C stretching), 1390.68, 1328.95, 1273.02, 1193.94, 1122.57, 1072.42, 1045.42 (C-H bending, C=S and C-N stretching, aromatic C-H in plane bending), 933.55, 867.97, 833.25, 761.88, 719.45, 688.59, 653.87 (Aromatic C-H out of plane bending and C-S stretching) (Supplementary Material Figure S89).
1H NMR (500 MHz, DMSO-d6): 6.56 (s, 1H), 7.39 (t, J = 7.10 Hz, 7.20 Hz, 14.30 Hz, 1H), 7.49 (t, J = 7.35 Hz, 7.45 Hz, 14.80 Hz, 2H), 7.75 (t, J = 8.75 Hz, 8.15 Hz, 16.90 Hz, 7H), 7.87 (d, J = 8.50 Hz, 2H), 8.03 (d, J = 7.95 Hz, 2H), 8.26 (s, 1H), 8.50 (s, 1H), 10.28 (s, 1H), 11.99 (s, 1H) (Supplementary Material Figures S90–S92).
13C NMR (125 MHz, DMSO-d6): 108.28 (CH), 118.49 (2CH), 127.19 (2CH), 127.31 (2CH), 127.33 (CH), 128.13 (2CH), 128.34 (CH), 128.78 (2CH), 129.49 (2CH), 133.62 (C), 137.41 (C), 137.63 (C), 139.85 (C), 141.36 (CH), 142.03 (C), 143.15 (CH), 176.51 (C) (Supplementary Material Figure S93).
HRMS (ESI) (m/z) [M + H]+ calcd. for C23H19N5S: 398.1434, found: 398.1737 (Supplementary Material Figure S94).

3.2. DPP-4 Inhibitor Screening Assay

DPP-4 Inhibitor Screening Assay Kit (Cayman Chemicals, Ann Arbor, MI, USA), a fluorescence-based method, is used to screen the DPP-4 inhibitory effects of compounds 2ao and sitagliptin (Cayman Chemicals, Ann Arbor, MI, USA) according to the manufacturer’s instructions. This assay kit allows DPP-4 measurement using Gly-Pro-Aminomethylcoumarin (AMC) as a fluorogenic substrate. The free AMC group is released through the cleavage of the peptide bond by DPP leading to fluorescence that can be examined using appropriate excitation and emission wavelengths. Briefly, the experiment was performed triplicate in a white flat bottom 96 well plate. Thirty microlitres of assay buffer, 10 µL of DPP-4 enzyme, 10 µL of dimethyl sulfoxide (DMSO), and 50 µL of DPP substrate were added to 100% initial activity wells. Forty microlitres of assay buffer, 10 µL of DMSO, and 50 µL of DPP substrate were added to background wells. Thirty microlitres of assay buffer, 10 µL of DPP-4 enzyme, 10 µL of sample, and 50 µL of DPP substrate were added to inhibitor wells. Sitagliptin was used as a positive control inhibitor. The plate was incubated for 30 min at 37 °C, and fluorescence measurements were analyzed using an excitation wavelength of 360/40 nm and an emission wavelength of 460/40 nm in a multi-mode plate reader (Synergy HTX S1LFA, BioTek Instruments, Winooski, VT, USA). All assays were performed in triplicate. The fluorescence of the background wells was subtracted from all other wells before the calculations. The per cent inhibition was determined by the following formula in equation 1:
Inhibition% = [(Initial activity − Inhibitor)/Initial Activity] × 100
First, all compounds were studied at the same concentration (100 µM) with DPP-4 Inhibitor Screening Assay Kit for activity pre-screening results. After the elimination of the lower potency compounds, the effective compounds were tested at five different concentrations with 1:10 dilutions (10−1 to 10−5 µM) to determine the IC50 values.

3.3. Cytotoxicity

3.3.1. Cell Culture

NIH/3T3 mouse embryonic fibroblast (normal) cells (ATCC® CRL-1658™) were grown in RPMI 1640 medium (Sigma-Aldrich, St. Louis, MO, USA) supplemented with 2 mM L-glutamine (Gibco, Paisley, UK), 10% fetal bovine serum (Gibco, Paisley, UK) and 1% penicillin/streptomycin (Gibco, Paisley, UK) at 37 °C in a humidified incubator with a 5% CO2 atmosphere. Compounds 2ao were dissolved in DMSO (Sigma-Aldrich, St. Louis, MO, USA) and diluted to required concentrations with a fresh medium. The control group (solvent control) was prepared with a medium containing 0.1% DMSO.

3.3.2. MTT Assay

MTT [(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (Sigma-Aldrich, St. Louis, MO, USA) assay is based on the reduction of MTT by the mitochondrial dehydrogenase of intact cells to a purple formazan product [37]. Yellow MTT is reduced to purple formazan in the mitochondria of viable cells. This reduction takes place only when mitochondrial reductase enzymes are active, and therefore, the conversion can be directly related to the number of viable cells [38].
In order to determine IC50 concentrations of compounds 2ao, cell viability was determined by MTT assay. Briefly, cells were grown in 96-well plates at a density of 5 × 103 cells per well and subjected to different concentrations (500, 250, 125, 62.5 and 31.25 µM) of the compounds. After 24 h incubation, the MTT solution was added to wells to reach a final concentration of 0.5 mg/mL. The cells were incubated for a further 4 h and the then-current medium was removed and 100 µL of DMSO solution was added. The absorbance values were measured at 540 nm using a Cytation 3 Cell Imaging Multi-Mode Reader (BioTek, Winooski, VT, USA). Cell survival rates were expressed as the percentage of the DMSO (0.1%) solvent control and IC50 concentrations were calculated according to the analysis result.

3.4. Statistical Analyses

All descriptive data were expressed as the mean±standard deviation. All statistical analysis was performed by using GraphPad Prism Software version 8.4.2. A multiple comparisons test was applied to compare the mean of each column with the mean of control column with the Dunnet test. Mean standard deviation was calculated for quantitative data. p values less than 0.05 (≤0.05) were considered to be statistically significant.

3.5. Molecular Docking Studies

The most active DPP-4 inhibitors in this series and sitagliptin were docked to the active site of DPP-4. Ligands were set to the physiological pH (pH = 7.4) at the protonation step and the crystal structure of DPP-4 was retrieved from Protein Data Bank server (PDB code: 4FFW). The structures of the compounds were submitted in the protein preparation module of Schrodinger’s Maestro molecular modelling package (Schrödinger Release 2016-2: Schrödinger, LLC, New York, NY, USA). In molecular docking simulations, Glide/XP docking protocols were applied for the prediction of topologies of the compounds in the active site of the target enzyme [36].

3.6. In Silico ADME Studies

The pharmacokinetic profiles of compounds 2ao were predicted using the QikProp module of Schrödinger’s Molecular modelling package (Schrödinger Release 2016-2: QikProp, Schrödinger, LLC., New York, NY, USA, 2016). According to Lipinski’s rule of five and Jorgensen’s rule of three, all compounds were evaluated for their drug-likeness and oral bioavailability.

4. Conclusions

In conclusion, new 4-[4-(1H-pyrazol-1-yl)phenyl]-1-arylidenethiosemicarbazides (2ao) were designed as DPP-4 inhibitors and synthesized efficiently through a two-step procedure. Compounds 2ao were investigated for their in vitro DPP-4 inhibitory potencies using a fluorescence-based assay. According to the results, compound 2f bearing the bromo substituent at the para position of the benzylidene moiety was found to possess the most pronounced inhibitory effect on DPP-4 in this series (IC50 = 1.266 ± 0.264 nM) when compared to sitagliptin (IC50 = 4.380 ± 0.319 nM). The cytotoxic effects of the compounds on NIH/3T3 mouse embryonic fibroblast (normal) cells were examined. The MTT assay pointed out the safety of compound 2f as a promising DPP-4 inhibitor. Molecular docking studies pointed out that compound 2f revealed high affinity to the active site of DPP-4 and established strong hydrophobic interactions with key residues very similar to sitagliptin. Based on the safety profile and good tolerance of current available DPP-4 inhibitors, according to both in vitro and in silico studies compound 2f stands out as a potential antidiabetic agent showing DPP-4 inhibition.

Supplementary Materials

The following are available online. Figure S1: IR spectrum of compound 2a, Figure S2: 1H NMR spectrum of compound 2a, Figure S3: 1H NMR spectrum of compound 2a with integral values, Figure S4: 1H NMR spectrum of compound 2a (6–12 ppm), Figure S5: 13C NMR spectrum of compound 2a, Figure S6: HRMS spectrum of compound 2a, Figure S7: IR spectrum of compound 2b, Figure S8: 1H NMR spectrum of compound 2b, Figure S9: 1H NMR spectrum of compound 2b with integral values, Figure S10: 1H NMR spectrum of compound 2b (6–12 ppm), Figure S11: 13C NMR spectrum of compound 2b, Figure S12: HRMS spectrum of compound 2b, Figure S13: IR spectrum of compound 2c, Figure S14: 1H NMR spectrum of compound 2c, Figure S15: 1H NMR spectrum of compound 2c with integral values, Figure S16: 1H NMR spectrum of compound 2c (6–12 ppm), Figure S17: 13C NMR spectrum of compound 2c, Figure S18: HRMS spectrum of compound 2c, Figure S19: IR spectrum of compound 2d, Figure S20: 1H NMR spectrum of compound 2d, Figure S21: 1H NMR spectrum of compound 2d with integral values, Figure S22: 1H NMR spectrum of compound 2d (6–12 ppm), Figure S23: 13C NMR spectrum of compound 2d, Figure S24: HRMS spectrum of compound 2d, Figure S25: IR spectrum of compound 2e, Figure S26: 1H NMR spectrum of compound 2e, Figure S27: 1H NMR spectrum of compound 2e with integral values, Figure S28: 1H NMR spectrum of compound 2e (6–12 ppm), Figure S29: 13C NMR spectrum of compound 2e, Figure S30: HRMS spectrum of compound 2e, Figure S31: IR spectrum of compound 2f, Figure S32: 1H NMR spectrum of compound 2f, Figure S33: 1H NMR spectrum of compound 2f with integral values, Figure S34: 1H NMR spectrum of compound 2f (6.6–8.8 ppm), Figure S35: 1H NMR spectrum of compound 2f (10.2–12.2 ppm), Figure S36: 13C NMR spectrum of compound 2f, Figure S37: HRMS spectrum of compound 2f, Figure S38: IR spectrum of compound 2g, Figure S39: 1H NMR spectrum of compound 2g, Figure S40: 1H NMR spectrum of compound 2g with integral values, Figure S41: 1H NMR spectrum of compound 2g (6–12 ppm), Figure S42: 13C NMR spectrum of compound 2g, Figure S43: HRMS spectrum of compound 2g, Figure S44: IR spectrum of compound 2h, Figure S45: 1H NMR spectrum of compound 2h, Figure S46: 1H NMR spectrum of compound 2h with integral values, Figure S47: 1H NMR spectrum of compound 2h (2.3–12 ppm), Figure S48: 13C NMR spectrum of compound 2h, Figure S49: HRMS spectrum of compound 2h, Figure S50: IR spectrum of compound 2i, Figure S51: 1H NMR spectrum of compound 2i, Figure S52: 1H NMR spectrum of compound 2i with integral values, Figure S53: 1H NMR spectrum of compound 2i (3.8–12 ppm), Figure S54: 13C NMR spectrum of compound 2i, Figure S55: HRMS spectrum of compound 2i, Figure S56: IR spectrum of compound 2j, Figure S57: 1H NMR spectrum of compound 2j, Figure S58: 1H NMR spectrum of compound 2j with integral values, Figure S59: 1H NMR spectrum of compound 2j (3.2–12 ppm), Figure S60: 13C NMR spectrum of compound 2j, Figure S61: HRMS spectrum of compound 2j, Figure S62: IR spectrum of compound 2k, Figure S63: 1H NMR spectrum of compound 2k, Figure S64: 1H NMR spectrum of compound 2k with integral values, Figure S65: 1H NMR spectrum of compound 2k (2.5–12 ppm), Figure S66: 13C NMR spectrum of compound 2k, Figure S67: HRMS spectrum of compound 2k, Figure S68: IR spectrum of compound 2l, Figure S69: 1H NMR spectrum of compound 2l, Figure S70: 1H NMR spectrum of compound 2l with integral values, Figure S71: 1H NMR spectrum of compound 2l (2.5–3.5 ppm), Figure S72: 1H NMR spectrum of compound 2l (6.5–12 ppm), Figure S73: 13C NMR spectrum of compound 2l, Figure S74: HRMS spectrum of compound 2l, Figure S75: IR spectrum of compound 2m, Figure S76: 1H NMR spectrum of compound 2m, Figure S77: 1H NMR spectrum of compound 2m with integral values, Figure S78: 1H NMR spectrum of compound 2m (1–3.3 ppm), Figure S79: 1H NMR spectrum of compound 2m (6.5–12 ppm), Figure S80: 13C NMR spectrum of compound 2m, Figure S81: HRMS spectrum of compound 2m, Figure S82: IR spectrum of compound 2n, Figure S83: 1H NMR spectrum of compound 2n, Figure S84: 1H NMR spectrum of compound 2n with integral values, Figure S85: 1H NMR spectrum of compound 2n (1–3.3 ppm), Figure S86: 1H NMR spectrum of compound 2n (6.5–12 ppm), Figure S87: 13C NMR spectrum of compound 2n, Figure S88: HRMS spectrum of compound 2n, Figure S89: IR spectrum of compound 2o, Figure S90: 1H NMR spectrum of compound 2o, Figure S91: 1H NMR spectrum of compound 2o with integral values, Figure S92: 1H NMR spectrum of compound 2o (6.5–12 ppm), Figure S93: 13C NMR spectrum of compound 2o, Figure S94: HRMS spectrum of compound 2o.

Author Contributions

Conceptualization, B.S. and M.D.A.; methodology, B.S., Ş.G., Z.C. and M.D.A.; software, B.S., H.S., Ş.G., Z.C. and M.D.A.; validation, B.S., Ş.G. and Z.C.; formal analysis, B.S., H.S., Ş.G. and Z.C.; investigation, B.S.; resources, B.S. and M.D.A.; data curation, B.S., Ş.G., Z.C. and M.D.A.; writing—original draft preparation, B.S.; writing—review and editing, B.S., H.S., Ş.G., Z.C. and M.D.A.; visualization, B.S. and Ş.G.; project administration, B.S. and M.D.A.; funding acquisition, M.D.A. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Anadolu University Scientific Research Projects Commission under the grant no: 2005S019 and 1905S050.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cantley, J.; Ashcroft, F.M. Q&A: Insulin secretion and type 2 diabetes: Why do β-cells fail? BMC Biol. 2015, 13, 33. [Google Scholar]
  2. Kerru, N.; Singh-Pillay, A.; Awolade, P.; Singh, P. Current anti-diabetic agents and their molecular targets: A review. Eur. J. Med. Chem. 2018, 152, 436–488. [Google Scholar] [CrossRef] [PubMed]
  3. Artasensi, A.; Pedretti, A.; Vistoli, G.; Fumagalli, L. Type 2 diabetes mellitus: A review of multi-target drugs. Molecules 2010, 163, 51–59. [Google Scholar] [CrossRef] [PubMed]
  4. Havale, S.H.; Pal, M. Medicinal chemistry approaches to the inhibition of dipeptidyl peptidase-4 for the treatment of type 2 diabetes. Bioorg. Med. Chem. 2009, 17, 1783–1802. [Google Scholar] [CrossRef]
  5. Thornberry, N.A.; Gallwitz, B. Mechanism of action of inhibitors of dipeptidyl-peptidase-4 (DPP-4). Best Pract. Res. Clin. Endocrinol. Metab. 2009, 23, 479–486. [Google Scholar] [CrossRef]
  6. Gilbert, M.P.; Pratley, R.E. Efficacy and safety of incretin-based therapies in patients with type 2 diabetes mellitus. Am. J. Med. 2009, 122, S11–S24. [Google Scholar] [CrossRef]
  7. Lambeir, A.M.; Scharpé, S.; de Meester, I. DPP4 inhibitors for diabetes—What next? Biochem. Pharmacol. 2008, 76, 1637–1643. [Google Scholar] [CrossRef]
  8. Patel, B.D.; Ghate, M.D. Recent approaches to medicinal chemistry and therapeutic potential of dipeptidyl peptidase-4 (DPP-4) inhibitors. Eur. J. Med. Chem. 2014, 74, 574–605. [Google Scholar] [CrossRef]
  9. Davies, M.J.; Bianchi, C.; del Prato, S. Use of incretin-based medications: What do current international recommendations suggest with respect to GLP-1 receptor agonists and DPP-4 inhibitors? Metabolism 2020, 107, 154242. [Google Scholar] [CrossRef]
  10. Yaribeygi, H.; Maleki, M.; Sathyapalan, T.; Jamialahmadi, T.; Sahebkar, A. Anti-inflammatory potentials of incretin-based therapies used in the management of diabetes. Life Sci. 2020, 241, 117152. [Google Scholar] [CrossRef]
  11. Matteucci, E.; Giampietro, O. Dipeptidyl peptidase-4 (CD26): Knowing the function before inhibiting the enzyme. Curr. Med. Chem. 2009, 16, 2943–2951. [Google Scholar] [CrossRef]
  12. Enz, N.; Vliegen, G.; de Meester, I.; Jungraithmayr, W. CD26/DPP4—A potential biomarker and target for cancer therapy. Pharmacol. Ther. 2019, 198, 135–159. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, H.; Guo, L.; Xing, J.; Li, P.; Sang, H.; Hu, X.; Du, Y.; Zhao, L.; Song, R.; Gu, H. The protective role of DPP4 inhibitors in atherosclerosis. Eur. J. Pharmacol. 2020, 875, 173037. [Google Scholar] [CrossRef] [PubMed]
  14. Mulakayala, N.; Reddy CH, U.; Iqbal, J.; Pal, M. Synthesis of dipeptidyl peptidase-4 inhibitors. Tetrahedron 2010, 66, 4919–4938. [Google Scholar] [CrossRef]
  15. Ahrén, B. DPP-4 inhibitors. Best Pract. Res. Clin. Endocrinol. Metab. 2007, 21, 517–533. [Google Scholar] [CrossRef] [PubMed]
  16. Deng, X.; Tavallaie, M.S.; Sun, R.; Wang, J.; Cai, Q.; Shen, J.; Lei, S.; Fu, L.; Jiang, F. Drug discovery approaches targeting the incretin pathway. Bioorg. Chem. 2020, 99, 103810. [Google Scholar] [CrossRef] [PubMed]
  17. Shao, S.; Xu, Q.; Yu, X.; Pan, R.; Chen, Y. Dipeptidyl peptidase 4 inhibitors and their potential immune modulatory functions. Pharmacol. Ther. 2020, 209, 107503. [Google Scholar] [CrossRef]
  18. Stoian, A.P.; Sachinidis, A.; Stoica, R.A.; Nikolic, D.; Patti, A.M.; Rizvi, A.A. The efficacy and safety of dipeptidyl peptidase-4 inhibitors compared to other oral glucose-lowering medications in the treatment of type 2 diabetes. Metabolism 2020, 109, 154295. [Google Scholar] [CrossRef]
  19. Yu, Y.; Kalinowski, D.S.; Kovacevic, Z.; Siafakas, A.R.; Jansson, P.J.; Stefani, C.; Lovejoy, D.B.; Sharpe, P.C.; Bernhardt, P.V.; Richardson, D.R. Thiosemicarbazones from the old to new: Iron chelators that are more than just ribonucleotide reductase inhibitors. J. Med. Chem. 2009, 52, 5271–5294. [Google Scholar] [CrossRef]
  20. Alexacou, K.M.; Tenchiu Deleanu, A.C.; Chrysina, E.D.; Charavgi, M.D.; Kostas, I.D.; Zographos, S.E.; Oikonomakos, N.G.; Leonidas, D.D. The binding of β-d-glucopyranosyl-thiosemicarbazone derivatives to glycogen phosphorylase: A new class of inhibitors. Bioorg. Med. Chem. 2010, 18, 7911–7922. [Google Scholar] [CrossRef]
  21. Kulkarni, N.V.; Ravenkar, V.K.; Kirasur, B.N.; Hugar, M.H. Transition metal complexes of thiosemicarbazones with quinoxaline hub: An emphasis on antidiabetic property. Med. Chem. Res. 2012, 21, 663–671. [Google Scholar] [CrossRef]
  22. Moorthy, N.S.; Cerqueira, N.M.; Ramos, M.J.; Fernandes, P.A. Aryl- and heteroaryl-thiosemicarbazone derivatives and their metal complexes: A pharmacological template. Recent Pat. Anti-Cancer Drug Discov. 2013, 8, 168–182. [Google Scholar] [CrossRef] [PubMed]
  23. Marques, K.M.R.; Desterro, M.R.D.; de Arruda, S.M.; Neto, L.N.D.A.; de Lima, M.D.C.A.; de Almeida, S.M.V.; da Silva, E.C.D.; de Aquino, T.M.; da Silva-Júnior, E.F.; Júnior, J.X.D.A.; et al. 5-Nitro-thiophene-thiosemicarbazone derivatives present antitumor activity mediated by apoptosis and DNA intercalation. Curr. Top. Med. Chem. 2019, 19, 1075–1091. [Google Scholar] [CrossRef] [PubMed]
  24. Shehzad, M.T.; Imran, A.; Njateng, G.S.S.; Hameed, A.; Islam, M.; Al-Rashida, M.; Uroos, M.; Asari, A.; Shafiq, Z.; Iqbal, J. Benzoxazinone-thiosemicarbazones as antidiabetic leads via aldose reductase inhibition: Synthesis, biological screening and molecular docking study. Bioorg. Chem. 2019, 87, 857–866. [Google Scholar] [CrossRef] [PubMed]
  25. Datar, P.A.; Jadhav, S.R. Development of pyrazole compounds as antidiabetic agent: A review. Lett. Drug Des. Discov. 2014, 11, 686–703. [Google Scholar] [CrossRef]
  26. Shu, S.; Cai, X.; Li, J.; Feng, Y.; Dai, A.; Wang, J.; Yang, D.; Wang, M.W.; Liu, H. Design, synthesis, structure-activity relationships, and docking studies of pyrazole-containing derivatives as a novel series of potent glucagon receptor antagonists. Bioorg. Med. Chem. 2016, 24, 2852–2863. [Google Scholar] [CrossRef] [PubMed]
  27. Faria, J.V.; Vegi, P.F.; Miguita, A.G.C.; dos Santos, M.S.; Boechat, N.; Bernardino, A.M.R. Recently reported biological activities of pyrazole compounds. Bioorg. Med. Chem. 2017, 25, 5891–5903. [Google Scholar] [CrossRef]
  28. Ganguly, S.; Jacob, S.K. Therapeutic outlook of pyrazole analogs: A mini review. Mini Rev. Med. Chem. 2017, 17, 959–983. [Google Scholar] [CrossRef]
  29. Karrouchi, K.; Radi, S.; Ramli, Y.; Taoufik, J.; Mabkhot, Y.N.; Al-aizari, F.A.; Ansar, M. Synthesis and pharmacological activities of pyrazole derivatives: A review. Molecules 2018, 23, 134. [Google Scholar] [CrossRef] [Green Version]
  30. Abdellatif, K.R.A.; Fadaly, W.A.A.; Kamel, G.M.; Elshaier, Y.A.M.M.; El-Magd, M.A. Design, synthesis, modeling studies and biological evaluation of thiazolidine derivatives containing pyrazole core as potential anti-diabetic PPAR-γ agonists and anti-inflammatory COX-2 selective inhibitors. Bioorg. Chem. 2019, 82, 86–99. [Google Scholar] [CrossRef]
  31. Singh, P.; Mothilal, S.; Kerru, N.; Singh-Pillay, A.; Gummidi, L.; Erukainure, O.L.; Islam, S. Comparative α-glucosidase and α-amylase inhibition studies of rhodamine-pyrazole conjugates and their simple rhodanine analogues. Med. Chem. Res. 2019, 28, 143–159. [Google Scholar] [CrossRef]
  32. Brullo, C.; Rapetti, F.; Bruno, O. Pyrazolyl-ureas as interesting scaffold in medicinal chemistry. Molecules 2020, 25, 3457. [Google Scholar] [CrossRef] [PubMed]
  33. Yoshida, T.; Akahoshi, F.; Sakashita, H.; Kitajima, H.; Nakamura, M.; Sonda, S.; Takeuchi, M.; Tanaka, Y.; Ueda, N.; Sekiguchi, S.; et al. Discovery and preclinical profile of teneligliptin (3-[(2S,4S)-4-[4-(3-methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl]pyrrolidin-2- ylcarbonyl]thiazolidine): A highly potent, selective, long-lasting and orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. Bioorg. Med. Chem. 2012, 20, 5705–5719. [Google Scholar]
  34. Hsu, T.; Chen, C.; Tsai, T.; Cheng, J.; Wu, S.; Chang, C.; Chien, C.; Yeh, K.; Huang, Y.; Huang, C.; et al. (1,3-Diphenyl-1H-pyrazol-4-yl)- methylamine analogues as inhibitors of dipeptidyl peptidases. J. Chin. Chem. Soc. 2009, 56, 1048–1055. [Google Scholar] [CrossRef]
  35. Wu, D.; Jin, F.; Lu, W.; Zhu, J.; Li, C.; Wang, W.; Tang, Y.; Jiang, H.; Huang, J.; Liu, G.; et al. Synthesis, structure–activity relationship, and pharmacophore modeling studies of pyrazole-3-carbohydrazone derivatives as dipeptidyl peptidase IV inhibitors. Chem. Biol. Drug Des. 2012, 79, 897–906. [Google Scholar] [CrossRef] [PubMed]
  36. Altıntop, M.D.; Temel, H.E.; Sever, B.; Çiftçi, G.A.; Kaplancikli, Z.A. Synthesis and evaluation of new benzodioxole-based thiosemicarbazone derivatives as potential antitumor agents. Molecules 2016, 21, 1598. [Google Scholar] [CrossRef] [Green Version]
  37. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 16, 55–63. [Google Scholar] [CrossRef]
  38. Özdemir, A.; Altıntop, M.D.; Sever, B.; Cantürk, Z.; Kaplancıkli, Z.A. Synthesis and evaluation of tetrazole-based hydrazone derivatives bearing a pyridine moiety as antimicrobial agents. Lett. Drug Des. Discov. 2015, 12, 687–693. [Google Scholar] [CrossRef]
Sample Availability: Samples of compounds 2ao are available from the authors.
Figure 1. Some DPP-4 inhibitors.
Figure 1. Some DPP-4 inhibitors.
Molecules 25 05003 g001
Figure 2. Some thiosemicarbazones endowed with antidiabetic activity.
Figure 2. Some thiosemicarbazones endowed with antidiabetic activity.
Molecules 25 05003 g002
Figure 3. Some pyrazole derivatives as DPP-4 inhibitors.
Figure 3. Some pyrazole derivatives as DPP-4 inhibitors.
Molecules 25 05003 g003
Scheme 1. The synthetic route for the preparation of compounds 2ao. Reagents and conditions: (i) NH2NH2·H2O, ethanol, rt, 4 h; (ii) ArCHO, ethanol, reflux, 8 h.
Scheme 1. The synthetic route for the preparation of compounds 2ao. Reagents and conditions: (i) NH2NH2·H2O, ethanol, rt, 4 h; (ii) ArCHO, ethanol, reflux, 8 h.
Molecules 25 05003 sch001
Figure 4. The diagram for NMR data of compounds 2ao.
Figure 4. The diagram for NMR data of compounds 2ao.
Molecules 25 05003 g004
Figure 5. Docking poses of compounds 2f, 2g, 2i, 2k and 2o along with sitagliptin in the active site of DPP-4 (Blue and green dashes: π-π interactions, yellow dashes: hydrogen bonding).
Figure 5. Docking poses of compounds 2f, 2g, 2i, 2k and 2o along with sitagliptin in the active site of DPP-4 (Blue and green dashes: π-π interactions, yellow dashes: hydrogen bonding).
Molecules 25 05003 g005
Figure 6. Docking poses of compounds 2f and 2g along with sitagliptin (A) and docking interactions of compounds 2f and 2g (B) in the active site of DPP-4 (Blue and green dashes: π-π interactions, yellow dashes: hydrogen bonding).
Figure 6. Docking poses of compounds 2f and 2g along with sitagliptin (A) and docking interactions of compounds 2f and 2g (B) in the active site of DPP-4 (Blue and green dashes: π-π interactions, yellow dashes: hydrogen bonding).
Molecules 25 05003 g006aMolecules 25 05003 g006b
Table 1. The percentage inhibition values of compounds 2ao and sitagliptin for DPP-4.
Table 1. The percentage inhibition values of compounds 2ao and sitagliptin for DPP-4.
Compound (100 µM)RDPP-4 Inhibition%
2aH20.67 ± 1.82
2bNO235.54 ± 1.75
2cCN28.04 ± 1.63
2dF27.28 ± 2.03
2eCl38.44 ± 2.14
2f *Br98.20 ± 0.89
2g *CF362.82 ± 1.19
2hCH353.16 ± 3.10
2i *OCH377.18 ± 2.21
2jSO2CH359.84 ± 1.13
2k *SCH370.44 ± 1.03
2lN(CH3)257.34 ± 1.04
2mCH(CH3)247.43 ± 1.29
2nC(CH3)328.64 ± 1.76
2o *C6H575.72 ± 1.48
Sitagliptin *-90.38 ± 2.17
* These compounds were selected for further analysis to calculate the IC50 values. IC50 calculation was not performed for the concentrations higher than 100 µM for the results below 40% inhibition, and the results above 60% inhibition were analyzed for concentrations less than 100 µM indicated by an asterisk.
Table 2. IC50 values of compounds 2f, 2g, 2i, 2k, and 2o for DPP-4 inhibition.
Table 2. IC50 values of compounds 2f, 2g, 2i, 2k, and 2o for DPP-4 inhibition.
CompoundIC50 (nM)
2f1.266 ± 0.264
2g4.775 ± 0.296
2i43.312 ± 0.372
2k22.671 ± 0.301
2o18.061 ± 0.311
Sitagliptin4.380 ± 0.319
Table 3. Predicted Absorption, Distribution, Metabolism, and Excretion (ADME) properties of compounds 2ao.
Table 3. Predicted Absorption, Distribution, Metabolism, and Excretion (ADME) properties of compounds 2ao.
CompoundQPlogPo/w *QPlogBB *CIQPlogS *QPlogKhsa *Human Oral Absorption% *
2a4.07−0.25−4.730.33100.00
2b3.38−1.40−5.250.3090.34
2c3.34−1.14−5.670.1994.39
2d4.31−0.14−5.080.37100.00
2e4.56−0.09−5.410.44100.00
2f4.64−0.08−6.300.46100.00
2g5.040.07−6.080.58100.00
2h4.38−0.27−5.000.48100.00
2i4.17−0.33−5.040.35100.00
2j2.86−1.25−4.77−0.0290.38
2k4.69−0.24−5.450.50100.00
2l4.50−0.37−5.210.49100.00
2m5.03−0.36−5.560.72100.00
2n5.30−0.35−5.840.85100.00
2o5.67−0.37−6.500.89100.00
* QPlogPo/w: Predicted octanol/water partition coefficient (−2.00 to 6.50), QPlogBB: brain/blood partition coefficient (−3.00 to 1.20), CIQPlogS: The conformation-independent predicted aqueous solubility (−6.50 to 0.50), QPlogKhsa: binding to human serum albumin (−1.50 to 1.50), Percent Human-Oral Absorption: human oral absorption on 0–100% scale (>80% is high, <25% is poor).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sever, B.; Soybir, H.; Görgülü, Ş.; Cantürk, Z.; Altıntop, M.D. Pyrazole Incorporated New Thiosemicarbazones: Design, Synthesis and Investigation of DPP-4 Inhibitory Effects. Molecules 2020, 25, 5003. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25215003

AMA Style

Sever B, Soybir H, Görgülü Ş, Cantürk Z, Altıntop MD. Pyrazole Incorporated New Thiosemicarbazones: Design, Synthesis and Investigation of DPP-4 Inhibitory Effects. Molecules. 2020; 25(21):5003. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25215003

Chicago/Turabian Style

Sever, Belgin, Hasan Soybir, Şennur Görgülü, Zerrin Cantürk, and Mehlika Dilek Altıntop. 2020. "Pyrazole Incorporated New Thiosemicarbazones: Design, Synthesis and Investigation of DPP-4 Inhibitory Effects" Molecules 25, no. 21: 5003. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25215003

Article Metrics

Back to TopTop