key: cord-0832248-1m8wmvl3 authors: Shehzad, Muhammad Tariq; Imran, Aqeel; Hameed, Abdul; Rashida, Mariya al; Bibi, Marium; Uroos, Maliha; Asari, Asnuzilawati; Iftikhar, Shafia; Mohamad, Habsah; Tahir, Muhammad Nawaz; Shafiq, Zahid; Iqbal, Jamshed title: Exploring synthetic and therapeutic prospects of new thiazoline derivatives as aldose reductase (ALR2) inhibitors date: 2021-05-11 journal: RSC advances DOI: 10.1039/d1ra01716k sha: 1b707cf1010de3d42d7e0ec15e9aaeb1cc089fe0 doc_id: 832248 cord_uid: 1m8wmvl3 Inhibition of aldose reductase (ALR2) by using small heterocyclic compounds provides a viable approach for the development of new antidiabetic agents. With our ongoing interest towards aldose reductase (ALR2) inhibition, we have synthesized and screened a series of thiazoline derivatives (5a–k, 6a–f, 7a–1 & 8a–j) to find a lead as a potential new antidiabetic agent. The bioactivity results showed the thiazoline-based compound 7b having a benzyl substituent and nitrophenyl substituent-bearing compound 8e were identified as the most potent molecules with IC(50) values of 1.39 ± 2.21 μM and 1.52 ± 0.78 μM respectively compared with the reference sorbinil with an IC(50) value of 3.14 ± 0.02 μM. Compound 7b with only 23.4% inhibition for ALR1 showed excellent selectivity for the targeted ALR2 to act as a potential lead for the development of new therapeutic agents for diabetic complications. The incidence of Diabetes mellitus (DM) disease is increasing alarmingly and more than 400 million people are affected all over the world. 1 Diabetes complications affect about 25% of the elderly population over the age of 65, and this proportion is steadily growing. 2 The majority of the population affected with DM belongs to under-developed or developing regions of the world. [3] [4] [5] [6] According to a recent study between COVID-19 and diabetes, the COVID-19 patients with diabetes have a two-fold higher risk of mortality and disease incidence than COVID-19 patients without diabetes [00] . In case of progression of this disease severe diabetic complications result such as neuropathy, nephropathy, mood disorders, diabetic retinopathy. These complications are generally a result of hyperglycemia 7 which initiates the polyol pathway due to non-insulin dependent glucose uptake. This pathway primarily involves NADPH dependent reduction of glucose to sorbitol, the enzyme responsible for this reduction is aldose reductase (AR; ALR2; EC 1. 1.1.21) belonging to aldo-keto reductase enzyme superfamily. The sorbitol in turn converts metabolically via another enzyme sorbitol dehydrogenase into fructose, resulting in increase in the glucose ux. 8 High glucose level in diabetes promotes the combination of glucose to ALR2 and metabolized about one third of the total glucose to sorbitol via the polyol pathway in tissues such as retina, lens, peripheral nerves and kidneys. As a result, the regulated polyol pathway accumulates the sorbitol in cells causing swelling of cell, osmotic imbalance and changes in permeability of membrane. Sorbitol does not penetrate the cellular membranes especially that of eye lens. Moreover, the drastic lessening of NAD + and NADPH alters the cellular redox potentials and weakens the enzymatic activities like that of glutathione reductase and nitric oxide synthase (NOS); worsening the intracellular oxidative stress level. Stress level is also increased via free radicals produced from a number of radical precursors like protein kinase C (PKC) isomer, advanced glycation end products (AGEs), poly-ADP-ribose polymerase (PARP) and mitogen-activated protein kinase (MAPK). High level of free radicals damages a number of tissues. Hence, all the oxidative stress reactions mediated by ALR2 along with the polyol pathway represent important pathogenesis of diabetic complications. 9 From aldo-keto reductase (AKR), those reducing the aldehydes are called aldehyde reductases (ALR1, EC 1.1.1.2) while those involved in the reduction of ketones are termed as ketoreductases (also belonging to AKR family). Both enzymes have almost similar structure differing just in their active sites. 10 The one accurately capable to reduce the aldehyde functionality of glucose in polyol pathway is ALR1. This enzyme is also involved in metabolism of 3-deoxyglucosone and methyl glyoxal causing toxic glycation end products. In contrast, it also assists the reductive detoxication of reactive aldehydes. An example is the reduction of aldehyde phospholipids to regulate the pro-inammatory response. 11 There are a number of studies reported the aldose reductase inhibitors (ARIs). [12] [13] [14] [15] [16] [17] Up to yet now, only one ARI drug; Epalrestat, ONO Pharmaceutical, Osaka, Japan has been marketed. 18, 19 Though the polyol pathway inhibition is more challenging to reduce the complications of diabetes; some isolated natural products have been used as potent ARIs. Some synthesized compounds have same active functionalities as that of potent natural product and have been entered in to clinical trials ( Fig. 1 ). It is of utmost importance to develop potent and selective ARIs (ALR1 and ALR2 share about 65% sequence homology), which can regulate the polyol pathway and combat secondary diabetic complications. 20, 21 The present work focuses on the synthesis of a series of novel thiazoline based inhibitors and their evaluation as ARIs. The compounds containing benzoxazinone, adamantyl, benzodioxane and indole nuclei have been synthesized and investigated for their diverse biological activities as many of these moieties are also the part of bioactive natural products. [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] Incorporation or conjugation of thiazoline moieties with another biologically important nucleus is expected to enhance their biological potential. In view of this and in search of novel bioactive molecules, the study was designed and aimed to prepare a number of various thiazolines possessing benzoxazinone, adamantyl, benzodioxane and indole moieties to evaluate their enzyme inhibition potential with an expectation that they may display more potent activity and thus result into the development of different compounds of medicinal interest. 2.1 Chemistry of thiazolines derivatives 5a-k, 6a-f, 7a-i and 8a-j In the current study, the thiazoline derivatives 5-8 were designed and prepared in variable yields (76-92%) by using four different types of carbonyl group bearing compounds 1 i.e. 6acetyl-2H-benzo[b] [1, 4] oxazin-3(4H)-one, 1-acetyl adamantane, 1,4-benzodioxan-6-yl methyl ketone and indole-3carboxaldehyde. The starting materials bearing carbonyl group were treated in equimolar quantities with N-substituted thiosemicarbazides 2 in methanol as solvent. The reaction was catalyzed by using glacial acetic acid as catalyst to get thiosemicarbazones 3 as intermediate. 34, 35 Further, the thiosemicarbazone derivatives 3 were reacted with a range of 4substituted 2-bromoacetophenones 4 in solvent ethanol along with sodium acetate. The resulting mixture was heated at reux till the complete consumption of starting material, monitored by TLC analysis. The pure product was obtained via recrystallization from absolute ethanol (Scheme 1). The structures of thiazoline derivatives (5a-k, 6a-f, 7a-i and 8a-j) were conrmed by using different spectroscopic techniques that include IR spectra, NMR spectroscopy and microanalysis (CHN). The infrared spectra of a typical thiazolines from 5a-k series showed a stretching band of NH group at 3184-3338 cm À1 , carbonyl group (C] O) of lactam moiety at 1663-1748 cm À1 and imine group C]N bands were appeared at 1578-1593 cm À1 regions consequently, the compounds in Scheme 1 Preparation of thiazoline derivatives 5a-k, 6a-f, 7a-i and 8a-j. Fig. 2 The X-ray crystal structure of thiazoline derivative 5h. Note: the ORTEP diagram of 5h drawn at 50% probability level with H-atoms as small circles of arbitrary radii. The minor disordered parts of chloroform are not shown for clarity. Fig. 3 The X-ray crystal structure of thiazoline derivative 7i. Note: the ORTEP diagram of 7i drawn at 50% probability level with H-atoms as small circles of arbitrary radii. The minor disordered part is not shown for clarity. thiazoline series 6a-f and 7a-i, showed imine group (C] N) in the range of 1558-1617 cm À1 . Furthermore, the indole-based thiazolines 8a-j, showed NH stretching band at 3125-3444 cm À1 while C]N bond in the range of 1603-1615 cm À1 . The proton 1 H-NMR spectra of different thiazoline series 5a-k and 8a-j, displayed broad singlets for lactam NH and indole NH The synthetic thiazoline derivatives (5a-k, 6a-f, 7a-1 & 8a-j) were tested against aldehyde reductase enzyme (ALR1), and their anti-diabetic potential by evaluating inhibitory activity against aldose reductase (ALR2). Results indicated that out of thirty six compounds tested, eight of them, 5f, 6a, 6b, 6c, 7g, 7h, 7i, and 8e were found active inhibitors of ALR2 and ALR1 enzymes (Table 2 ). However, compound 6d, 6e, 6f, 7a, 7b, 7c and 8f were identied as selective ALR2 inhibitors (Fig. 4 ). Compound 5f, one of the 2H-1,4-benzoxazin-3(4H)-one bearing derivatives were active against ALR1 and ALR2 having IC 50 value of 3.13 AE 1.45 mM and 3.24 AE 2.72 mM, respectively. The substitution of nitrophenyl with bromophenyl or chlorophenyl, as in compound 5b and 5j, showed weak activity against both ARL1 and ARL2 enzymes in comparison to sorbinil and valproic acid with respective IC 50 values of 3.14 AE 0.02 mM and 57.4 AE 0.89 mM ( Table 2 ). In general, compounds (6a-6f) containing adamantane substituent demonstrated the most promising activity among all the derivatives. Out of six, three compounds 6d, 6e and 6f showed a good inhibitory activity and selectively against ALR2 with IC 50 values 4.21 AE 2.35 mM, 2.18 AE 0.83 mM and 3.51 AE 2.31 mM respectively. Compounds 6a, 6b and 6c were also found to be active against ALR1 and ALR2 enzymes (Fig. 5) . Among the series, compound 7b showed high inhibition potential against ALR2 (IC 50 ¼ 1.39 AE 2.21 mM). However, compound 7b was found to have considerably selective activity against ARL2 exhibiting only 23.4% inhibition against ALR1. The inhibition potential of chlorophenyl substituted thiazoline derivative 7i against ALR2 was much lower than the aforementioned compound with IC 50 of 38.2 AE 1.43 mM. Furthermore, an improved inhibitor potency of compound 7i was also observed against ALR1 (IC 50 4.01 AE 0.39 mM). Among the indolyl substituted thiazoline derivatives, compound 8e having a nitrophenyl moiety showed high inhibitory potency against ARL2 and ARL1, with IC 50 values of 1.52 AE 0.78 mM and 2.94 AE 1.34 mM respectively. However, the inhibitory activity of chlorophenyl substituted thiazoline derivative 8h, was weakened for both enzymes ALR2 and ALR1 demonstrating 10.32% and 33.4% inhibition respectively. Furthermore, the compound 8f was a selective ALR2 inhibitor than ALR1 exhibiting only 12.4% inhibition (Fig. 6 ). The other indolyl substituted compounds (8a, 8b, 8c, 8d, 8g, 8h, 8i, & 8j) were found inactive with less than 50% inhibitory activity against ALR2 as well against ARL1 enzymes. To rationalize the mode of binding and nature of binding site interactions, molecular docking studies were carried out using BioSolveIT's LeadIT soware. 36 For each inhibitor, the top 10 docked conformations were further evaluated for their binding free energy using HYDE utility (part of LeadIT soware), the conformation with most favorable binding free energy was retained for further analysis The crystal structures of porcine ALR1 (ref. 37) and human ALR2 (ref. 38) were downloaded from the Protein Data Bank [PDB ids: 3FX4 at 1.99Å and 1US0 at 0.66 A respectively]. 39 Docking protocol was validated by re-docking of the co-crystallized ligand. The docking protocol was able to reproduce the experimentally bound conformation of cocrystallized ligand (FX4) with an rmsd of 1.03. For docking against ALR1, three of the most active inhibitors 7h, 7i and 8e were selected. All compounds were found to bind in the same area of the binding pocket as the co-crystallized inhibitor FX4 (Fig. 7) . By analyzing the binding site interactions of the cocrystallized ligand (ALR1 inhibitor), FX4 ([5-(3carboxymethoxy-4-methoxybenzylidene)-2,4-dioxothiazolidin-3yl]acetic acid) it can be seen that the amino acid residues that are important for binding are Arg312, Phe298, Trp220, Trp22, Arg309, and Ala219. When docking studies of ALR1 inhibitors (7h, 7i, 8e) were carried out, same amino acids were found to be involved in binding these inhibitors (Table S1 †). Fig. S1 † shows the docked conformation of compound 7h. The oxygen atom of the benzodioxane ring was making hydrogen bond with Met302. The nitrogen atom of the C]N moiety next to the thiazole ring was making a hydrogen bond with Arg312. The OH group of Tyr50 was acting as a hydrogen bond donor towards the uorine atom. The carbonyl oxygen atom of Tyr 50 was acting as a halogen bond acceptor towards the chlorine atom. A number of hydrophobic interactions were also observed. Phe125 was making pi-pi stacked interactions with the thiazole and the phenyl ring attached to thiazole ring. Ile299 was making alkyl and pi-alkyl interactions with the methyl group and the phenyl ring of benzodioxane ring respectively. Ile49 was making pi-alkyl interactions with both chlorophenyl and uorophenyl rings, whereas Trp114 was making pi-alkyl interaction with the methyl group. Docking of compound 7i revealed hydrogen bonded interactions between the oxygen atom of the benzodioxane ring and Val300, and between the nitrogen atom of the C]N group and Met302 (Fig. S2 †) . A number of hydrophobic interactions were also observed. Ile299 was making alkyl and pi-alkyl interactions with the methyl group and phenyl ring of benzodioxane ring respectively. Pro301 was making also alkyl interaction with the methyl group. Lys23 was alkyl interaction with the chlorine atom and pi-alkyl interaction with chlorophenyl ring. Arg218 was making pi-alkyl interaction with the phenyl ring of benzodioxane ring, whereas Ala219 was making pi-alkyl interaction with the thiazole ring. Docking of compound 8e revealed two hydrogen bonds of Arg309 and Arg312 with nitrogen atom of C]N bond and oxygen atom of the nitro group (Fig. S3 †) . Notable hydrophobic interactions include pi-sigma and pi-alkyl interaction of Ile49 with indole phenyl ring and pyrrole ring of indole respectively. Tyr50 was making pi-pi stacked interaction with pyrrole ring of indole. Ile299 and Met302 were making pi-alkyl interactions with thiazole ring and nitro phenyl ring respectively. Docking studies of ALR2 inhibitors were also carried out for most active inhibitors 6e, 7b and 8e. Prior to docking, the docking protocol was veried by re-docking the co-crystallized ligand LDT ({2-[(4-bromo-2-uorobenzyl)carbamothioyl]-5-uorophenoxy}acetic acid) from the ALR2 (PDB id: 1su0). The docking protocol was able to reproduce the experimentally observed conformation of LDT with rmsd of <2. Moreover all compounds were found to bind at the same region of the active site as that of the co-crystallized inhibitor LDT (Fig. S4 †) . Docked conformation of compound 6e is shown in Fig. S5 . † The nitrogen atom of the C]N group was making a hydrogen bond with Trp20. A number of hydrophobic interactions were observed that are deemed necessary for efficient binding. Leu300 was making a pi-sigma interaction with the chloro phenyl ring. Trp20 was making pi-pi stacked and pi-alkyl interactions with the benzyl ring and the methyl group respectively. Trp111 was making a pi-stacked interaction with the chloro phenyl ring and a pi-alkyl interaction with the chloro group. Two pi-sulfur interactions were also observed. Trp219 was making a pi-sulfur contact with the sulfur atom of the thiazole ring, whereas the sulfur atom of Cys298 was making pisulfur contact with the benzyl ring. For compound 7b (Fig. 8) , similar interactions were observed. Trp20 was within hydrogen bond distance (1.99Å and 2.12Å) of both nitrogen atoms of C]N groups. Hydrophobic interactions include Trp111 making a pi-pi stacked interaction with the bromo phenyl ring and a pi-alkyl interaction with the bromine atom. Trp20 was making a pi-pi T-shaped interaction with the thiazole ring. Pro218 was making a pi-alkyl interaction with the methyl group. Leu300 was making pi-alkyl interaction with both benzyl and bromo phenyl ring, whereas Val47 was making pi-alkyl interaction with the thiazole ring. A pi-sulfur interaction was also observed between Tyr48 and the sulfur atom of thiazole ring. Docking of compound 8e was also carried out, its docked conformation along with binding site interactions are shown in Fig. 9 . Both oxygen atoms of the nitro group were making hydrogen bonds with Trp111 and Tyr48, His110 is also within hydrogen bond distance to the nitro group. It is important to note that Trp111, Tyr48 and His110 are the same amino acids that are involved in binding the carboxylate group of standard inhibitor LDT. Leu300 was making hydrophobic interactions, pi-sigma and pi-alkyl with pyrrole ring of indole, and the phenyl indole ring respectively. Phe122 was making pi-pi T-shaped interaction with both thiazole and indole rings. Another pi-pi T-shaped interaction was observed between Trp20 and nitro phenyl ring. Moreover, an intramolecular pi-pi T shaped contact was also observed between the nitro phenyl and benzyl ring, this orientation may additionally stabilize the binding of inhibitor. The nitro phenyl ring was also found to be involved in a pi-alkyl interaction with Val47. An electrostatic attractive interaction was observed between Lys77 and the oxygen atom of the nitro group. Another electrostatic (pi-anion) interaction was observed between Trp20 and same oxygen atom of the nitro group. The conformational stabilities of the apoenzymes (ALR1 and ALR2) and their protein + ligand complexes, both cognate and test compounds, were performed to simulate protein exibility. The structure of proteins (apoproteins) were rst subjected to MD run of 50 ns and then docked poses of cognate ligands and selected ligands (holoenzymes) were submitted to MD run for 50 ns. The time evolution of the radius of gyration of 3FX4 and 1US0, apoenzymes and holoenzymes, exposed in the applied electric elds are shown in Fig. S6 † and 10 , respectively. The graph indicating that the average value of R g slightly uctuating between 1.92-1.97 nm for ALR1 (3FX4), while for ALR2 (1US0), the radius of gyration lies between 1.89 and 1.93 nm, signifying a ne degree of compactness. The average values of RMSD over a period 50 ns for 3FX4 and 1US0, apoenzymes and holoenzymes, exhibited minute uctuations, reaffirming stable complex formation between enzymes and test compounds, as shown in Fig. S7 † and 11 , respectively. RMSF analyzes the portions of structure that are uctuating from their mean structure the most (or least). The (RMSF) of ALR1 and ALR2 were examined, it was observed that residues were found stable in both the cases (Fig. S8, † and 12 ). In this research, we have synthesized thiazoline derivatives (5ak, 6a-f, 7a-1 & 8a-j), which were tested against aldehyde reductase (ALR1), and aldose reductase (ALR2) enzymes to study their anti-diabetic potential. The results demonstrated that compounds containing adamantyl substituent (6a-6f) have most promising activity among all the derivatives. The compound 7b (with benzyl substituent) among the series was found signicantly selective against ARL2 with IC 50 value of 1.39 AE 2.21 mM compare to sorbinil, a reference inhibitor, with IC 50 values of 3.14 AE 0.02 mM. Furthermore, the compounds 6e also showed potency against ALR2 with IC 50 values of 2.18 AE 0.83 mM, whilst 6f presented slightly higher with IC 50 value of 3.51 AE 2.31 mM when compared with standard sorbinil. The compound 8e (with nitrophenyl substituent) demonstrated high potency and selectivity against ALR2 enzyme with IC 50 values of 1.52 AE 0.78 mM. In silico molecular docking study was also performed to further study the putative binding of active compounds with the target enzyme to nd lead compound for further steps of drug development. 6 Experimental section 6.1 General procedure for the synthesis of thiosemicarbazones (3) A solution of corresponding aldehyde or ketone 1 (6-acetyl-2Hbenzo[b] [1, 4] oxazin-3(4H)-one, 1-acetyl adamantane, 1,4benzodioxan-6-yl methyl ketone and indole-3-carboxaldehyde; 0.01 mol) in methanol (10 mL) was added to a hot stirred solution of appropriate N 4 -substituted thiosemicarbazide 2 (0.01 mol) in methanol (10 mL) . Aer adding few drops of glacial acetic acid as catalyst, the reaction mixture was heated under reux for 2-6 h. Upon completion of reaction, monitored through TLC, the hot reaction mixture was cooled to room temperature. The solid product obtained in each case was ltered, washed several times with hot methanol and dried under vacuum to afford the desired thiosemicarbazones 3 in pure form. The resultant thiosemicarbazone derivatives were used as such in the next step without any further purication. 6.2 General procedure for the synthesis of thiazoline derivatives (5) (6) (7) (8) A mixture of equimolar amounts of appropriate thiosemicarbazone derivative 3 (0.005 mol), 4-substituted (bromo, nitro, chloro) phenacyl bromide 4 (0.005 mol) and anhydrous sodium acetate (0.005 mol) in absolute ethanol (25 mL) was heated under reux with continuous stirring for 12-24 h. The reaction mixture was then partially concentrated on a rotary evaporator and le overnight. The precipitate formed in each case was ltered off, washed with warm diethyl ether, dried and recrystallized from absolute ethanol to furnish the target thiazoline derivatives 5-8 in pure form. The different compounds are characterized as under: [1, 4] oxazin-3(4H)-one (5a). : C, 57.81; H, 3.69; N, 10.79; found: C, 57.88; H, 3.65; N, 10.85. 6-((E)-1-((Z)-(4-(4-bromophenyl)-3-(3-methoxyphenyl)thiazol-2(3H)-ylidene) hydrazono) ethyl)-2H-benzo[b] [1, 4] .70 (s, 1H, CH-S), 6.75-6.77 (m, 1H, Ar-H), 6.86-6.88 (dd, 1H, J ¼ 0. 8 Hz, 8.4 Hz, 2H, 7.14 (d, 2H, J ¼ 2.0 Hz, 7.25 (t, 1H, J ¼ 8.0 Hz, 7.34 (dd, 1H, J ¼ 2.4 Hz, 8.8 Hz, 6-((E)-1-((Z)- (4-(4-bromophenyl)-3-(2,6-dimethylphenyl) thiazol-2(3H)-ylidene) hydrazono) ethyl)-2H-benzo[b] [1, 4] 6.35 (s, 1H, 3H, 2H, 1H, 2H, 1H, 7.54 (d, 1H, J ¼ 2.0 Hz, 7.86 (s, 1H, NH) ; 13 : C, 59.23; H, 4.23; N, 10.23; found: C, 59.30; H, 4.25; N, 10.20. 6- [1, 4] oxazin-3(4H)-one (5d). C, 61.85; H, 3.94; N, 11.42; found: C, 61.80; H, 3.98; N, 11.36. 6-((E)-1-((Z)-(3-(3-methoxyphenyl)-4-(4-nitrophenyl)thiazol-2(3H)-ylidene) hydrazono)ethyl)-2H-benzo[b] [1, 4] 2H, 7.79 (s, 1H, 2H, 2H, , 10.82 (s, 1H, NH) ; 13 C, 60.57; H, 4.11; N, 13.58; found: C, 60.51; H, 4.15; N, 13.52. 6-((E)-1-((Z)-(3-(4-uorophenyl)-4-(4-nitrophenyl)thiazol-2(3H)-ylidene) hydrazono)ethyl)-2H-benzo[b] [1, 4] C, 63.14; H, 4.51; N, 13.64; found: C, 63.18; H, 4.54; N, 13.57. 6-((E)-1-((Z)-(4-(4-chlorophenyl)-3-phenylthiazol-2(3H)-ylidene)hydrazono) ethyl)-2H-benzo[b] [1, 4] oxazin-3(4H)-one (5i). -methoxyphenyl)thiazol-2(3H)-ylidene) hydrazono) ethyl)-2H-benzo[b][1,4]oxazin-3(4H)-one (5j) 82 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 14.89 (CH 3 ), 55.43 (OCH 3 ), 67.25 (O-CH 2 ) -uorophenyl)thiazol-2(3H)-ylidene) hydrazono) ethyl)-2H-benzo[b][1,4]oxazin-3(4H)-one (5k) Ar-C), 113.89 (Ar-C), 115.34 (Ar-C), 116.31 (Ar-C) -(adamantan-1-yl)ethylidene)hydrazono)-4-(4-bromophenyl)-3-phenyl-2,3-dihydrothiazole (6a) 400 MHz) d ppm: 1.67-1.75 (m, 6H, adamantane -CH 2 ), 1.81 (m, 6H, adamantane -CH 2 ), 1.85 (s, 3H 94 (adamantane-C), 40.38 (adamantane-C) -(adamantan-1-yl)ethylidene)hydrazono)-4-(4-bromophenyl)-3-(naphthalen-1-yl)-2,3-dihydrothiazole (6b) 18 (s, 1H, CH-S), 6.89 (dd, 2H, J ¼ 2.0 Hz, 6.8 Hz, Ar-H), 7.13 (dd, 2H, J ¼ 2.0 Hz, 6.8 Hz, Ar-H), 7.22 (dd 400 MHz) d ppm: 1.56 (bs, 6H, CH 3 -C]N, CH 3 -Ar), 1.66-1.72 (m, 6H, adamantane -CH 2 -(adamantan-1-yl)ethylidene)hydrazono)-4-(4-bromophenyl m, 6H, adamantane -CH 2 ), 1.77-1.80 (bs, 6H, adamantane -CH 2 ), 2.00-2.02 (bs, 3H, adamantane -CH 2 ), 2.15 (s, 3H, CH 3 -Ar), 6.13 (s, 1H, CH-S 100 MHz) d ppm: 12.17 (CH 3 ) (m, 6H, adamantane -CH 2 ), 1.81 (s, 3H 26 (adamantane-C), 36.90 (adamantane-C) -(adamantan-1-yl)ethylidene)hydrazono)-4-(4-chlorophenyl)-3-(2,6-dimethyl phenyl bs, 6H, adamantane -CH 2 ), 2.00 (bs, 3H, adamantane -CH 2 ), 2.13 (s, 6H, 2ÂCH 3 -Ar), 6.08 (s, 1H, CH-S ] dioxin-6-yl) ethylidene) hydrazono)-3-phenyl-2,3-dihydrothiazole (7a) Ar-H) m, 4H, Ar-H); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 14.45 (CH 3 ), 49.30 (CH 2 ), 64.34 (O-CH 2 ] dioxin-6-yl) ethylidene) hydrazono)-3-(2,6-dimethylphenyl)-2,3-dihydrothiazole (7c) Ar-H) ]dioxin-6-yl)ethylidene) hydrazono)-3-(4-uorophenyl)-4-(4-nitrophenyl)-2,3-dihydrothiazole (7d) 12 (dd, 2H, J ¼ 2.0 Hz, 7.2 Hz, Ar-H); 13 C NMR (DMSOd 6 , 100 MHz) d ppm: 14 12 (s, 2H, -CH 2 -), 6.66 (s, 1H, CH-S), 6.88 (d, 1H, J ¼ 8.8 Hz, Ar-H), 7.02 (d, 2H, J ¼ 7.2 Hz ]dioxin-6-yl)ethylidene) hydrazono)-3-(2,6-dimethylphenyl)-4-(4-nitrophenyl)-2,3-dihydrothiazole (7f) 37 (dd, 2H, J ¼ 2.0 Hz, 7.2 Hz, Ar-H), 8.08 (dd, 2H, J ¼ 2.0 Hz, 7.2 Hz, Ar-H); 13 C NMR (DMSOd 6 , 100 MHz) d ppm: 14.63 (CH 3 ), 18.22 (2ÂCH 3 -Ar) ] dioxin-6-yl)ethylidene) hydrazono)-3-phenyl-2,3-dihydrothiazole (7g) 39 (d, 1H, J ¼ 2.0 Hz, Ar-H) ] dioxin-6-yl)ethylidene) hydrazono)-3-(4-uorophenyl)-2,3-dihydrothiazole (7h) 13 (s, 1H, CH-S), 6.82 (d, 1H, J ¼ 8.4 Hz Ar-C), 115.62 (Ar-C) 25 (s, 4H, -OCH 2 CH 2 O-), 5.00 (s, 2H, -CH 2 -), 5.92 (s, 1H, CH-S), 6.82 (d, 1H, J ¼ 8.4 Hz N, 8.83; found Z)-2-((E)-((1H-indol-3-yl)methylene)hydrazono)-4-(4-bromophenyl)-3-phenyl-2,3-dihydrothiazole (8a 36 (s, 1H, indole CH), 11.51 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 102 )methylene)hydrazono)-4-(4-bromophenyl)-3-cyclohexyl-2,3-dihydrothiazole (8b) C; IR y max (cm À1 ): 3125 (N-H) m, 1H, Ar-H), 8.54 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 25.09 (cyclohexyl-C 1H-indol-3-yl)methylene)hydrazono)-3-(4-uorophenyl)-4-(4-nitrophenyl)-2,3-dihydrothiazole (8c) MHz) d ppm: 6.93 (s, 1H, CH-S), 7.18-7.28 (m, 4H, Ar-H), 7.39-7.48 (m, 5H, CH]N, Ar-H), 7.72 (d, 1H, J ¼ 2.4 Hz 49 (Ar-C), 123.07 (Ar-C), 124.01 (Ar-C) yl)methylene)hydrazono)-4-(4-nitrophenyl)-3-phenyl-2,3-dihydrothiazole (8d) MHz) d ppm: 6.93 (s, 1H, CH-S), 7.18-7.22 (m, 2H, Ar-H), 7.33-7.35 (m, 3H, Ar-H), 7.39-7.46 (m, 5H 100 MHz) d ppm: 105 Ar-C) Z)-2-((E)-((1H-indol-3-yl)methylene)hydrazono)-3-benzyl-4-(4-nitrophenyl)-2,3-dihydrothiazole (8e) .46 (m, 1H, Ar-H) 53 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 48 indol-3-yl)methylene)hydrazono)-3-(3-methoxyphenyl)-4-(4-nitrophenyl Ar-H), 8.12 (d, 2H, J ¼ 8.8 Hz, Ar-H), 8.26-8.28 (m, 1H, Ar-H), 8.41 (s, 1H, indole CH), 11.55 (s, 1H, NH) Z)-2-((E)-((1H-indol-3-yl)methylene)hydrazono)-4-(4-chlorophenyl)-3-phenyl-2,3-dihydrothiazole (8g Ar-H), 8.27-8.30 (m, 1H, Ar-H), 8.36 (s, 1H, indole CH), 11.52 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 102 Yield 86%; m Ar-C) (m, 1H, Ar-H), 8.36 (s, 1H, indole CH), 11.53 (s, 1H, NH); 13 C NMR (DMSOd 6 , 100 MHz) d ppm: 102 (1H-indol-3-yl)methylene)hydrazono)-4-(4-chlorophenyl)-3-(2,6-dimethyl phenyl cm À1 ): 3392 (N-H) 72 (s, 1H 50 (s, 1H, NH); 13 C NMR (DMSO-d 6 , 100 MHz) d ppm: 18.25 (2ÂCH 3 -Ar) Treatment strategies against diabetes: Success so far and challenges ahead Association, 12. Older adults: standards of medical care in diabetes-2019 The global implications of diabetes and cancer IDF diabetes atlas: global estimates of the prevalence of diabetes for Identication of new potent inhibitor of aldose reductase from Ocimum basilicum Structure optimization of tetrahydropyridoindole-based aldose reductase inhibitors improved their efficacy and selectivity Exploration of thioxothiazolidinone-sulfonate conjugates as a new class of aldehyde/aldose reductase inhibitors: A synthetic and computational investigation Synthesis and structure-activity relationship studies of phenolic hydroxyl derivatives based on quinoxalinone as aldose reductase inhibitors with antioxidant activity Selectivity determinants of the aldose and aldehyde reductase inhibitor-binding sites Benzothiazolyl substituted iminothiazolidinones and benzamido-oxothiazolidines as potent and partly selective aldose reductase inhibitors Some new thiazolyl thiazolidinedione derivatives as aldose reductase inhibitors Pyrrolylbenzothiazole derivatives as aldose reductase inhibitors Determination of the inhibition proles of pyrazolyl-thiazole derivatives against aldose reductase and a-glycosidase and molecular docking studies Design, synthesis, in vitro and in silico investigation of aldose reductase inhibitory effects of new thiazole-based compounds Synthesis and aldose reductase inhibitory effect of some new hydrazinecarbothioamides and 4-thiazolidinones bearing an imidazo [2, 1-b] thiazole moiety, Turk An extensive research on aldose reductase inhibitory effects of new 4H-1, 2, 4-triazole derivatives Recent advances in aldose reductase inhibitors: potential agents for the treatment of diabetic complications Permeability characteristics of novel aldose reductase inhibitors using rat jejunum in vitro Benzoxazinone-thiosemicarbazones as antidiabetic leads via aldose reductase inhibition: Synthesis, biological screening and molecular docking study Addressing selectivity issues of aldose reductase 2 inhibitors for the management of diabetic complications Benzoxazinoids: reactivity and modes of action of a versatile class of plant chemical defenses Synthesis and antimicrobial activity of some benzoxazinoids derivatives of 2-nitrophenol and 3-hydroxy-2-nitropyridine Synthesis, structure-activity relationships studies of benzoxazinone derivatives as achymotrypsin inhibitors Design and synthesis of bioactive adamantane spiro heterocycles Design and synthesis of bioactive adamantanaminoalcohols and adamantanamines Synthesis and biology of 1, 4-benzodioxane lignan natural products Enantioselective synthesis, stereochemical correction, and biological investigation of the rodgersinine family of 1, 4-benzodioxane neolignans Synthesis and evaluation of new indole-based chalcones as potential antiinammatory agents Blood pressure-lowering efficacy of reserpine for primary hypertension Cumulative effects of reserpine on the pituitary-adrenocortical and sympathetic nervous system Distribution of reserpine in Rauvola species from India-HPTLC and LC-MS studies Al-Harrasi and Z. Shaq, Synthesis, characterization and molecular docking of some novel hydrazonothiazolines as urease inhibitors Exploring antidiabetic potential of adamantyl-thiosemicarbazones via aldose reductase (ALR2) inhibition Synthesis and urease inhibitory activity of 1, 4-benzodioxane-based thiosemicarbazones: Biochemical and computational approach Breakthrough: Chloroquine phosphate has shown apparent efficacy in treatment of COVID-19 associated pneumonia in clinical studies Structure of aldehyde reductase in ternary complex with a 5-arylidene-2, 4-thiazolidinedione aldose reductase inhibitor Ultrahigh resolution drug design I: Details of interactions in human aldose reductase-inhibitor complex at 0.66Å RCSB Protein Data Bank Acknowledgements Z. S. is thankful to Higher Education Commission, Islamabad, Pakistan through Project No. NRPU/6975 for nancial support. The authors declare no conict of interest.