Accepted Manuscript 2-Aminothiazole Derivatives as antimycobacterial agents: Synthesis, characterization, in vitro and in silico studies Parameshwar Makam, Tharanikkarasu Kannan PII:

S0223-5234(14)00914-3

DOI:

10.1016/j.ejmech.2014.09.086

Reference:

EJMECH 7394

To appear in:

European Journal of Medicinal Chemistry

Received Date: 21 February 2014 Revised Date:

21 June 2014

Accepted Date: 9 September 2014

Please cite this article as: P. Makam, T. Kannan, 2-Aminothiazole Derivatives as antimycobacterial agents: Synthesis, characterization, in vitro and in silico studies, European Journal of Medicinal Chemistry (2014), doi: 10.1016/j.ejmech.2014.09.086. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Graphical abstract 2-Aminothiazole derivatives as antimycobacterial agents: Synthesis, characterization, in

TE D

M AN U

SC

RI PT

vitro and in silico studies

A library of 2-aminothiazole derivatives were designed, synthesized and evaluated against

AC C

EP

Mycobacterium tuberculosis, H37Rv.

ACCEPTED MANUSCRIPT 2-Aminothiazole derivatives as antimycobacterial agents: Synthesis, characterization, in

2

vitro and in silico studies

3

Parameshwar Makam, Tharanikkarasu Kannan*

4

Department of Chemistry, Pondicherry University, Puducherry-605 014, India.

5

*

6

E -mail address: [email protected]; [email protected]

7

Abstract: A series of 2-aminothiazole derivatives with a wide range of substitutions at 2-, 4-

8

and 5- positions were designed and synthesized using Hantzsch thiazole synthesis. These

9

compounds were evaluated for their inhibitory potential against Mycobacterium tuberculosis

10

(Mtb), H37Rv. The compound, 7n showed high antimycobacterial activity with MIC value of

11

6.25 µM and the succeeding compounds, 7b, 7e and 7f also exhibited antimycobacterial

12

activity with MIC value of 12.50 µM. Docking studies of these molecules with β-Ketoacyl-

13

ACP Synthase (KasA) protein of Mtb have been carried out to understand the mechanism of

14

antimycobacterial action. The compound, 7n showed good interaction with KasA protein

15

with the Ki value of 0.44 µM.

16

Key words: Mycobacterium tuberculosis; tuberculosis; 2-aminothiazole; drug like molecule;

17

β-Ketoacyl-ACP Synthase; docking studies

EP

TE D

M AN U

SC

Corresponding author. Tel: +91-413-265 4708; Fax: +91-413-265 6740.

AC C

18

RI PT

1

1

ACCEPTED MANUSCRIPT 19 20

1. Introduction Tuberculosis (TB) is one of the world’s leading airborne contagious diseases that is mainly caused by Mycobacterium tuberculosis (Mtb), a species of genus Mycobacterium.

22

According to the eighteenth global TB report of the World Health Organization (WHO), it

23

was estimated that 1.3 million people died out of 8.6 million new infections in 2012 [1].

24

Although the death rate declined 45 % between 1990 and 2012, TB is next only to HIV/AIDS

25

in killing the highest numbers of people through single infectious disease. Whilst TB is an

26

ancient disease, its pathogen, Mtb was discovered in 1882 and the chemotherapy for this

27

disease was started in 1944 using streptomycin [2]. In the following years, the first line drugs

28

such as isoniazid, ethambutol, rifampicin, etc., and the second line drugs such as

29

ethionamide, p-amino salicylic acid, cycloserine, kanamycin, etc., have been used to treat TB

30

either independently or in combination with other drugs [3]. But, after 1950s, the rate in

31

developing novel and effective drugs was slow [4-6]. As a result, there is no sufficient

32

number of anti-tuberculosis drugs in the market. Due to this, TB patients still depend on the

33

drugs discovered in 1950s [7]. In addition, the constant emergence of multi, extensively and

34

totally drug – resistant strains of Mtb, and close synergy with HIV/AIDS [8] have made the

35

goal of wiping out TB from this world difficult. Poverty, complexity of the disease,

36

prolonged and poor administration of drugs are added reasons for the resurgence of TB as the

37

major global health burden [9-12], particularly in high TB burden countries [13].

39

SC

M AN U

TE D

38

RI PT

21

To find out a solution to this alarming increase of TB infections, many attempts have been made to understand the reasons behind the evolution and existence of resistant strains of

41

Mtb [14-16]. On the other hand, the generation and high-throughput screening of large

42

chemical entities with a wide spectrum of known and novel scaffolds have also been carried

43

out to identify lead anti-TB molecules with high efficacy, unique and novel mode of action.

44

[7, 17-20]. Among the diverse and medicinally relevant scaffolds, 2-aminothiazole

45

derivatives have exhibited good activity against Mtb, H37Rv [17, 21, 22]. Interestingly, 2-

46

aminothiazole derivatives are found to be structurally similar to thiolactomycin (TLM) [23] ,

47

a β-Ketoacyl-ACP Synthase (KasA) protein inhibitor [24]. TLM inhibits the biosynthesis of

48

mycolic acid, an essential cell wall component of Mtb, by inhibiting KasA protein [25].

49

Hence, it is expected that 2-aminothiazole derivatives may also inhibit growth of Mtb by

50

inhibiting biosynthesis of mycolic acid. 2-Aminothiazole derivatives have been validated

51

recently against UDP-galactopyranose mutase thwart to inhibit Mtb and found to have

AC C

EP

40

2

ACCEPTED MANUSCRIPT improved activities more than their parent thiazolidinone derivatives [26]. Nitazoxanide [27,

53

28], 2-aminothiazole-4-carboxylates and 2-amino-4-(2-pyridyl) thiazoles are other molecules

54

of this class now in clinical evaluations [19]. In addition to these, 2-aminothiazole scaffold is

55

attracting medicinal chemists continuously by being an integral part of drugs which are in

56

clinical practice for the treatment against tumor and cancer [29, 30], hypertension [31],

57

inflammation [32], analgesics [33], hypnotics [34], schizophrenia [35], HIV/AIDS [36],

58

malaria [37, 38] and microbial infections [39], to mention a few.

59 60

RI PT

52

Limited exploration of 2-aminothiazole derivatives against Mtb and the possibility of generating novel and potent Mtb inhibitors from this class encouraged us. In our previous

62

studies, we have synthesized novel 2-(2-hydrazinyl) thiazole derivatives comprising 2-

63

aminothiazole scaffold and evaluated for their inhibitory potentials against Mtb, H37Rv [40]

64

and Plasmodium falciparum, NF54 [41]. The good results obtained in these studies

65

encouraged us to go on to synthesize novel 2-aminothiazole derivatives. Hence, in the present

66

study, novel 2-aminothiazole derivatives are designed with a wide range of substitutions at

67

2-, 4- and 5- positions and evaluated their inhibitory potentials against Mtb by in vitro assays.

68

In addition, with the help of ligand – protein interaction studies through docking studies, an

69

insight into the mechanism of action has also been carried out against KasA protein.

70 71

2. Results and Discussion

72

75

EP

74

2.1. Rational design

The physico-chemical properties play a vital role in defining drug like molecule

AC C

73

TE D

M AN U

SC

61

76

(DLM) character and pharmacokinetics properties such as absorption, distribution,

77

metabolism, excretion and toxicity (ADMET) of a molecule [42, 43]. Certainly, the necessary

78

physico - chemical properties and their importance in the early drug discovery are best

79

explicated by the works of Lipinski [44] and others [43, 45, 46]. These rules state that the

80

molecule with a molecular mass less than 500, Log P value less than 5, hydrogen bond

81

donors less than 5, hydrogen bond acceptors less than 10, polar surface area less than 140 and

82

the number of rotatable bonds less than 10 could be a molecule with DLM character.

83

3

ACCEPTED MANUSCRIPT 84

In our previous investigation, 2-(2-hydrazinyl) thiazole derivatives have been designed without violating rule of five and synthesized. The synthesized compounds have

86

been evaluated for their inhibitory potential against Mtb, H37Rv and found that some of these

87

compounds show inhibitory potential against Mtb, H37Rv. As most of the anti-TB and

88

antibacterial compounds are exceptions to the rule of five [20, 45], in the present

89

investigation, we intentionally increased the lipophilicity of 2-aminothiazole derivatives by

90

designing compounds with the Log P values beyond 5. While designing, the structural

91

features of reported anti-TB molecules have been considered as shown in Figure 1. An

92

expectation during the designing is that the molecules with a defined optimal range of the

93

lipophilicity could influence in identifying molecules with high therapeutic success by

94

monitoring the ADMET properties of the molecules [47-49]. Bedaquiline, the only anti-TB

95

drug that has been approved by FDA in the last 40 years for drug-resistant tuberculosis

96

treatment is also a lipophilic drug [20]. Hence, by considering the cell wall permeability and

97

the solubility, during designing of 2- aminothiazole derivatives, core 2-aminothiazole

98

scaffold has been retained and additionally, substituted phenyl, naphthalene and positional

99

isomeric pyridyl systems have been incorporated on either side of 2-aminothiazole, i.e., at

M AN U

SC

RI PT

85

both 2- and 4- positions of the thiazole ring to induce lipophilicity and hydrophilicity

101

correspondingly. In addition to this, the H at 5- position has been replaced with other

102

functional groups such as CH3 and Br to understand the effect of this position on

103

antimycobacterial activity.

105



The physico – chemical properties of the designed 2-aminothiazole derivatives have

EP

104

TE D

100

been determined and the results are summarized in Table 1. The data reveal that all the

107

compounds follow the Lipinski rule of five except Log P values. As mentioned earlier, Log P

108

values of N-phenyl thiazol-2-amine derivatives (7a-7e, 7g, 7i-j and 7q-7s) and N-2-pyridyl

109

thiazol-2-amine derivatives (8c-j) have been intentionally maintained higher than 5. In

110

addition to these, the other parameters such as polar surface area and number of rotatable

111

bonds are found to be in the range of 24.919 - 83.63 and 3 - 6 respectively that are in the

112

range of recommended values.

113 114

AC C

106

2.2. Synthesis of 2-aminothiazole derivatives

115 116 117

The designed 2-aminothiazole derivatives have been synthesized according to the synthetic pathways described in Scheme 1. First, arylthioureas were synthesized by refluxing 4

ACCEPTED MANUSCRIPT corresponding anilines with ammonium thiocyanate in the presence dil. HCl [50] as shown in

119

Scheme 1 (i). But, pyridyl thioureas could not be achieved with the same procedure and

120

hence, an alternative procedure was used [51]. In this procedure, first, 2-pyridylamine,

121

ammonium thiocyanate and benzoyl chloride were reacted in acetone to yield N-(pyridin-2-

122

ylcarbamothioyl) benzamide. This was further hydrolyzed with 10% NaOH solution to yield

123

the corresponding pyridyl thioureas [51] as shown in Scheme 1 (ii). The phenyl and the

124

pyridyl thioureas have been characterized by the spectral methods and the data were found to

125

be in good agreement with the literature data [50-52].

127

SC

126

RI PT

118

In the next step, pyridyl α-bromo ketones have been synthesized from corresponding acetyl pyridines [51] as shown in Scheme 1 (iii). Finally, the aryl or pyridyl thioureas have

129

been heterocyclized to thiazole by reacting with the aryl or pyridyl α-halo ketones in the

130

presence of ethanol using the classical Hantzsch thiazole procedure [51] as shown in Scheme

131

1 (iv). All the synthesized compounds have been characterized using nuclear magnetic

132

resonance (NMR), infra-red (IR) and mass spectroscopic techniques. The aromatic protons

133

present in the pyridyl and phenyl systems are observed at 6.5 – 9 ppm. The appearance of the

134

characteristic peak of thiazole ring H at around 6.50 ppm and the disappearance of the

135

characteristic peaks of reactants in the 1H NMR spectra confirm the formation of the thiazole

136

ring. The characteristic peak of the NH at the 2nd position of the thiazole ring resonates at

137

around the 10.5 to 12 ppm. Similarly, the precise matching of the experimental mass and

138

CHNS data with the calculated data further confirms the formation of analytically pure

139

desired products. All the synthesized 2-aminothiazole derivatives have been classified into

140

four groups based on the structural similarities of the substitutions at the 2nd and 4th positions

141

on the 2-aminothiazole scaffold as shown in Table 2. In the first class of compounds,

142

substituted phenyl ring systems are attached on both 2nd and 4th positions on the 2-

143

aminothiazole scaffold. The spectral data of the compounds, 7a [53], 7b [53], 7f [54] 7g [55,

144

56] and 7h [55] are in agreement with the literature reports. In the second class, the

145

substituted 2-pyridyl and phenyl systems are incorporated on the 2nd and 4th positions of the

146

2-aminothiazole scaffold respectively. Among this class of compounds, the compounds, 7l

147

[51], 7o [51, 55] and 7p [55] are reported and the data is in agreement with the reported

148

literature. Similarly, in the third class of compounds, substituted pyridyl and phenyl systems

149

are incorporated on the 4th and 2nd positions of the 2-aminothiazole scaffold respectively.

150

The fourth class of compounds includes substituted pyridyl systems on both 2nd and 4th

AC C

EP

TE D

M AN U

128

5

ACCEPTED MANUSCRIPT 151

positions of the 2-aminothiazole scaffold. The spectral data of the reported compounds, 8l

152

[57] and 8n [58] are in agreement with the literature.

153

155

2.3. Single crystal and crystal packing studies The spatial arrangement of the drug molecules is an important factor and its

RI PT

154

understanding is more helpful in designing and identifying the better hit and lead drug

157

molecules with enhanced biological activity [51, 59, 60]. The spatial arrangement of the

158

molecules in its stable conformation can be identified by the single crystal X-ray studies. The

159

present 2-aminothiazole derivatives contain commonly three aromatic cyclic systems in their

160

structures viz. substituted phenyl, thiazole and pyridyl systems. Hence, based on the structural

161

diversity of the molecules, 7g and 8l have been selected as the representative molecules of 2-

162

aminothiazole derivatives. The molecule 7g contains both phenyl rings attached to 2nd and 4th

163

positions of 2-aminothiazole ring and the molecule 8l contains both pyridyl rings attached to

164

the 2nd and 4th positions of the 2-aminothiazole scaffold. The single crystal structures and

165

crystal packing diagrams of 7g and 8l have been solved and the results are summarized in

166

Table 3.

168

M AN U

TE D

167

SC

156



The single crystal structure of 7g is represented in the ORTEP diagram with 40% ellipsoid probability and its crystal packing is depicted in Figure 2. The single crystal

170

structure reveals that 7g is stabilized by its monohydrate form i.e., solvation by one water

171

molecule. The alignment of the three planar rings of the molecule, the 4-methoxy phenyl ring

172

systems attached to 4th positions of thiazole ring and the 2-aminothiazole ring are oriented

173

almost in the same plane. Whereas, the 4-hydroxy phenyl system attached to NH of the 2-

174

aminothiazole ring is perpendicular to the 2-aminothiazole ring (Figure 2a). The single

175

crystals are oriented in the opposite directions i.e., head to tail dimer fashion and are

176

stabilised by inter molecular hydrogen bonds. There are two inter molecular hydrogen

177

bonding (2.284 Å, 159.98°) exist between -NH group present in the 2-aminothiazole ring of

178

one molecule and the N present in the thiazole ring of other molecule and vice versa. In

179

addition, π – π interactions also exist between two 4-hydroxy phenyl rings (2.844 Å, 2.915 Å)

180

present in 7g. The solvent, water molecule is sandwiched between the two 7g molecules and

181

it is stabilising the packing of the crystals by forming four inter molecular hydrogen bond

182

interactions (2.05 Å, 126.51°; 2.04 Å, 126.41°) with two 7g molecules. The interaction

AC C

EP

169

6

ACCEPTED MANUSCRIPT 183

between Cl atom present in 7g and H2O (2.844 Å, 139.05°) also exits. All the hydrogen

184

bonding interactions and the orientation of the molecules in the crystal lattice are shown in

185

Figure 2b and 2c along the c- and a-axis respectively. The single helix arrangement can also

186

be visualised from the a-axis is shown in Figure 2c.

187

The single crystal structure of 8l reveals that the three thiazole ring and the pyridyl

RI PT

188

ring systems attached to 2nd and 4th positions are aligned in same plane. Hence, the molecule

190

is planar as shown in the single crystal structure in ORTEP diagram with 40% ellipsoid

191

probability (Figure 3a). The crystal packing reveals that the two molecules of 8l are

192

stabilised by the two intermolecular hydrogen bonds (2.101 Å, 167.91°) between the NH

193

group present in the 2nd position of the thiazole ring and the N present in the 3-pydidyl ring

194

and vice versa. The crystal packing of the molecules with two intermolecular hydrogen bonds

195

along a- and b-axis is shown in Figure 3 b and c respectively. The alignment of 8l molecules

196

in the layered and single helical fashion can be viewed along the a-axis as shown in Figure

197

3b.

M AN U

SC

189



198 199

2.4. Antimycobacterial activity and structure activity relationship (SAR) studies

TE D

200

After successful completion of design, synthesis and structural confirmation, these

202

four classes of 2-aminothiazole derivatives have been evaluated for their antimycobacterial

203

activities by Resazurin microtiter assay (REMA), an in vitro assay against Mtb, H37Rv [40]. It

204

is interesting to note that anti-TB activity results are varied based on the substitutions at 2nd,

205

4th and 5th positions of thiazole ring. In the first category of compounds, firstly, 4-fluoro

206

phenyl attached in the 2nd positions of the 2-aminothiazole scaffold kept constant and the

207

substitutions at 4th position and 5th positions have been varied. The variations at 4th position

208

i.e., 4-chloro phenyl system (7a), exhibits good activity with MIC value 25 µM. When the

209

chlorine is replaced with bromine (7b), the potency increases to one fold with MIC value of

210

12.5 µM. Interestingly, when the H at 5th position of the 7b is replaced with the methyl group

211

(7c) and bromine (7d), the potentency drops down to the MIC values of 25 µM and 100 µM

212

respectively. This clearly indicates the importance of the 5th position and its scope in

213

identifying the successful candidates. Similarly, when the 4-bromo phenyl of 7b is replaced

214

with 4-methoxy phenyl (7f) and naphthalene, the potential is retained with 12.5 µM. To

215

understand the effect of phenyl derivatives attached on the 2nd positions of the 2-

216

aminothiazole scaffold, 4-hydroxy and 2-nitro- 4-chloro phenyl derivatives have been

AC C

EP

201

7

ACCEPTED MANUSCRIPT 217

prepared. When the fluorine present in the compounds 7a and 7f is replaced with 4-hydroxy

218

group, the compounds 7g and 7h loses their potentials and exhibit MIC values 100 µM and

219

200 µM respectively. Similarly, the compounds 7i and 7j, the 2-nitro- 4-chloro phenyl

220

replaced derivatives of 7f and 7e show no activity with MIC value of >200 µM and this result

221

shows the importance of fluorine at 4th position. In the second category of compounds, the substituted phenyl systems at 2nd position

RI PT

222

are same and phenyl systems at 4th position are replaced with pyridyl systems. Keeping the 3-

224

pyridyl system intact at 4th position and varying the 2nd position with 4-flouro (7k), 4-bromo

225

(7m), 4-hydroxy (7o) and 2-nitro- 4-chloro (7r) phenyl systems lead to formation of the

226

compounds with no antimycobacterial activity (>200 µM). Interestingly, when 3-pyridyl

227

system is replaced with 4-pyridyl system, the activity enhances in the case of 4-flouro (7l)

228

and 4-bromo (7n) with MIC values 25 µM and 6.25 µM respectively. But, the compounds

229

with 4-hydroxy (7p) and 2-nitro- 4-chloro (7s) phenyl systems show no activity. The 2-

230

pyridyl systems with 2-nitro- 4-chloro phenyl (7q) do not make any difference in the activity.

M AN U

231

SC

223

In the third class of compounds, the 2-pyridyl group is introduced at the 2nd position of the 2-aminothiazole. In addition to this, substituted phenyl systems at 4th position and

233

methyl or bromine at 5th position have also been introduced to investigate the effect of 2-

234

pyridyl system. Unfortunately, all these compounds have exhibited no inhibition with MIC

235

value >200 µM against the Mtb, H37Rv. Similarly, to understand the effect pyridine systems

236

on both sides of 2-aminothiazole scaffold, fourth category of compounds have been

237

synthesized by keeping 3- or 4-pyridyl system at 4th position and 2-pyridyl derivative of NH

238

at 2nd position. In this category, 8m and 8n inhibit Mtb with MIC value 100 µM and rest of

239

the compounds are inactive with MIC value >200 µM.

EP

Overall, 4-flourophenyl system at 2nd position and H at 5th position of 2-aminothiazole

AC C

240

TE D

232

241

are enhancing the activity in the first class of compounds. Similarly, in the second class of

242

compounds, 4-flouro and 4-bromophenyl system on the 2nd position and 4-pyridyl systems on

243

the 4th position are enhancing the activity. The structural changes in the third and fourth class

244

of compounds did not influence much on enhancing the antimycobacterial activity. The

245

structure activity relationship studies are summarised in the Figure 4. The effect of Log P on

246

inhibition of Mtb also analysed for all the 34 compounds and the results are summarised in

247

Figure 5. When the Log P value is increased from 3.08 to 3.46 (8n, 8f) there is no difference

248

in antimycobacterial activity, but, when it increases to 3.71 (7l) the activity also increased.

249

When the log P value is further increased to 4.33 (7g) the antimycobacterial activity

250

decreased significantly. Further increase of Log P value up to 4.77 (7m, 7f) increases the 8

ACCEPTED MANUSCRIPT 251

activity enormously. After this, the activity reduced when log P value reaches 5.09 (7e).

252

From 5.54 to 6.35 (7d, 7a, 7i and 7b) the antimycobacterial activity is very good. Further

253

increase from 6.35 to 6.64 (7c) decreases the activity. Overall, the antimycobacterial activity

254

of 2-aminothiazole was found to be good when the Log P value is from 4.25 to 4.75 and from

255

5.5 to 6.5.

257

RI PT



256

2.5. Docking studies with KasA protein

258 259

The positive in vitro results encouraged us to study their possible mode of action through in silico methods using docking studies. As 2-aminothiazole derivatives are

261

structurally similar to TLM and TLM is potential inhibitor of the KasA protein of Mtb, in the

262

present study, KasA protein has been chosen as receptor target. KasA protein plays a vital

263

role in the biosynthesis of mycolic acid, long chain fatty acids and the essential components

264

of the Mtb cell wall [24]. Hence among all the 2-aminothiazole derivatives, the compounds,

265

7b, 7n, 8a and 8n have been chosen as the representative compounds based on their in vitro

266

activities and structural variations.

M AN U

267

SC

260

As anticipated, the selected compounds have exhibited good interactions and inhibition constant (Ki) against KasA protein. The compounds, 7b, 7n, 8a and 8n have shown

269

the Ki values of 1.28 µM, 0.44 µM, 30.20 µM and 30.19 µM with a binding energy of -8.04

270

Kcal/mol, -8.66 Kcal/mol, -11.66 Kcal/mol and -11.81 Kcal/mol respectively. The inhibition

271

of the protein by 7n is attained through hydrogen bonding interactions of N present in the 4-

272

pyridyl ring present at the 4th position of the 2-aminothiazole ring with COOH of the

273

phenylalanine (PHE) – 402. In addition to this, the NH present at the 2nd position of the 2-

274

aminothiazole ring has shown the hydrogen bond interactions with threonine (THR) – 315

275

and THR – 313 residues as shown in Figure 6. Hence, from the correlation of the results

276

obtained by in vitro and in silico analyses, it may be concluded that the present molecules

277

could be targeting the KasA protein and in turn disturbing the cell wall biosynthesis by

278

obstructing mycolic acid synthesis.

AC C

EP

TE D

268



279 280

3. Conclusion

281 282

The designed 2-aminothiazole derivatives with variations in the Log P values have

283

been designed and synthesized. All the synthesized compounds have been evaluated for their

284

inhibitory potentials against Mtb, H37Rv and their SAR studies have been carried out. Among 9

ACCEPTED MANUSCRIPT all the compounds, 4-flourophenyl group at 2nd position of the 2-aminothiazole ring system

286

has been proven to be responsible for contributing the activity. The compound 7n has shown

287

the highest inhibition with the MIC values of 6.25 µM and 0.44 µM by in vitro and in silico

288

analyses respectively. The continuation of the efforts in identifying the new molecules with

289

special focus on the present lead compounds to achieve improved antimycobacterial activities

290

are under progress in our laboratory.

291

4.

292

4.1. Materials and methods

Experimental

The substituted anilines, 2-, 3- or 4- acetyl pyridines, benzoyl chloride and aryl α –

SC

293

RI PT

285

haloketones were purchased either from Sigma–Aldrich, USA or Himedia Biosciences or

295

Spectrochem Pvt. Ltd (Mumbai, India). The chemicals procured were used without further

296

purification. The spectral data obtained by 1H NMR and 13C NMR were recorded with 400

297

MHz and 101 MHz Bruker Avance-II NMR instrument. Thermo Nicolet 6700 FT-IR

298

spectrometer was used to record the IR spectra and only major peaks are reported in cm-1.

299

Similarly, Elementar Vario EL-II CHNS analyzer was used for the Elemental analysis of all

300

the compounds. Chemical ionization (CI) or negative-ion electrospray ionization (ESI)

301

method was used to record the mass spectra (MS) by Thermo Scientific High Resolution

302

Magnetic Sector MS DFS. For the analysis of single crystals, Oxford diffractometer was used

303

and CrysAlis PRO, Oxford Diffraction Ltd., Version 1.171.34.44 (release 25-10-2010

304

CrysAlis171. NET) software has been used for the data collection, cell refinement and data

305

reduction purposes. The olex2.solve (compiled Oct 25 2010, 18:11:34) and SHELXL have

306

been used for solving the structure and to refine single crystal respectively. ChemBioDraw

307

Ultra has been used to deduce all the IUPAC names for the synthesized compounds.

TE D

EP

AC C

308

M AN U

294

309 310 311

4.2. General procedure for the synthesis of phenylthioureas (1a-d)

312

anilines (0.1 mol) were added and warmed to get a clear solution. Ammonium thiocyanate

313

(0.11 mol) dissolved in water (15 mL) was gradually added to this solution. The reaction

314

mixture was refluxed till the completion of the reaction and then more than the half of the

315

volume of the water present in the reaction mixture was distilled off. The phenyl thiourea was

To the dilute hydrochloric acid (15 mL) solution, the corresponding substituted

10

ACCEPTED MANUSCRIPT 316

obtained on gradual cooling of the reaction mixture, and it was filtered, washed with water

317

and recrystallized from ethanol.

318 319 320 321

4.3. General procedure for the synthesis of pyridylthioureas (3a-c)

322

chloride (65 mmol) was added dropwise by maintaining temperature between 20 – 25 °C.

323

Then the temperature of reaction mixture was raised to reflux temperature and stirred for 15

324

to 30 min. Then, the reaction mixture was cooled to 25 °C and appropriate pyridyl amine (64

325

mmol) was added gradually. The mixture was stirred at reflux temperature for a further 30

326

min, cooled to 20 °C, and poured on to ice. The mixture was stirred for another 30 min., and

327

then the precipitate was filtered, washed with water (15 mL) and dried. The solid was added

328

to a solution of NaOH (10% w/v, 100 mL) at 80 °C and continued stirring for 30 min, then

329

cooled to 20 °C and poured onto ice/HCl (6 M, 50 mL) solution. The pH of the mixture was

330

adjusted to 10 using conc. NH3 solution and stirred for 30 min. The precipitate was filtered

331

and washed with water (20 mL) and dried. The precipitate was purified by recrystallization

332

from ethanolic solution to afford the desired product in 85-95% yield.

M AN U

SC

RI PT

In dry acetone (50 mL), NH4SCN (69 mmol) was taken and to this solution, benzoyl

333

335 336

4.4. General procedure for the synthesis of (bromoacetyl)pyridines (5a-c)

TE D

334

Corresponding acetylpyridine (80 mmol) was taken in 30 % HBr/AcOH (100 mL) and to this stirred solution, Br2 (80 mmol) was added dropwise at 15 °C. The mixture was stirred

338

at 40 °C for 1 h and then at 75 °C for 1 h. Then, the reaction mixture was cooled to 20 °C.

339

The cooled reaction mixture was diluted with Et2O (400 mL), and continued to stir for 30

340

min. The precipitate obtained was filtered, washed with Et2O (25 mL), and dried under

341

vacuum to give the (bromoacetyl)pyridines as the hydrobromide salt [51].

AC C

342

EP

337

343 344 345

4.5. General procedure for the synthesis of titled 2-aminothiazole analogues (7a-8o)

346

solution, corresponding α-bromoacetylpyridine (1 mmol) or α-haloacetophenone (1 mmol)

347

derivative was added at 25- 30 °C. The reaction mixture was refluxed for 3-5 h or until the

348

reaction was complete as evidenced using TLC. Then, the reaction mixture was gradually

349

poured into ice-water (20 mL) and stirred for another 30 min. To this, aqueous 1N Na2CO3

350

solution was added to neutralise and to maintain pH ∼ 8. The precipitated aminothiazole

The appropriate thiourea (1 mmol) was dissolved in ethanol (∼10 mL) and to this

11

ACCEPTED MANUSCRIPT 351

derivatives were filtered and collected. The crude 2-aminothiazoles were purified by

352

recrystallization from ethanol.

353 354

4.5.1. 4-(4-Chlorophenyl)-N-(4-fluorophenyl)thiazol-2-amine (7a) White solid from EtOH (88% yield). m.p. 170.3-171.5 °C; 1H NMR (400 MHz,

356

CDCl3): δ 7.75 – 7.73 (m, 1H), 7.48 – 7.36 (m, 2H), 7.13 (dd, J = 9.1, 8.1 Hz, 1H), 6.70 (s,

357

1H), 3.40 (s, 1H);

358

132.46, 129.58, 127.58, 123.68, 122.73, 122.64, 117.06, 117.06, 116.83, 116.83, 104.05; IR

359

(KBr): 3255, 3066, 1607, 1505, 1404, 1238, 1091, 841, 769 cm-1. Anal. Calcd. for

360

C15H10ClFN2S: C, 59.11; H, 3.31; N, 9.19; S, 10.52; found: C, 59.13; H, 3.37; N, 9.22; S,

361

10.53; HR-MS (ESI+): m/z [M + H]

362

02330.

C NMR (101 MHz, DMSO-d6): δ 167.43, 159.22, 145.12, 134.49,

364

Calcd. for C15H10ClFN2S: 304.02326; found: 304.

M AN U

363

+

SC

13

RI PT

355

4.5.2. 4-(4-Bromophenyl)-N-(4-fluorophenyl)thiazol-2-amine (7b) White solid from EtOH (86% yield). m.p. 179.5 – 180.5 °C; 1H NMR (400 MHz,

366

CDCl3): δ 7.67 (d, J = 8.7 Hz, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.40 (dd, J = 9.1, 4.5 Hz, 1H),

367

7.12 (dd, J = 9.1, 8.1 Hz, 1H), 6.72 (s, 1H). 13C NMR (101 MHz, CDCl3): δ 167.43, 159.22,

368

145.12, 134.49, 132.46, 129.58, 127.58, 123.68, 122.73, 122.64, 117.06, 116.83, 100.55; IR

369

(KBr): 3254, 3065, 1606, 1504, 1401, 1238, 1006, 840, 729 cm-1. Anal. Calcd. for

370

C15H10BrFN2S: C, 51.59; H, 2.89; N, 8.02; S, 9.18; found: C, 51.57; H, 2.92; N, 8.04; S, 9.21;

371

HR-MS (ESI+): m/z [M + H] + Calcd. for C15H10BrFN2S: 347.97320; found: 347.97322.

TE D

365

373

4.5.3

EP

372

4-(4-Bromophenyl)-N-(4-fluorophenyl)-5-methylthiazol-2-amine (7c) White solid from EtOH (88% yield). m.p. 252.6-254°C; 1H NMR (400 MHz, DMSO-

375

d6): δ 7.67 (d, J = 8.7 Hz, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.40 (dd, J = 9.1, 4.5 Hz, 1H), 7.12

376

(dd, J = 9.1, 8.1 Hz, 1H), 6.72 (s, 1H), 2.45(s, 3H);

377

160.96, 158.81, 156.40, 141.84, 137.03, 132.91, 131.42, 130.25, 120.83, 119.88, 117.12,

378

115.65, 11.94; IR (KBr): 3448, 3074, 1621, 1594, 1506, 1230, 1002, 826, 645 cm-1. Anal.

379

Calcd. for C16H12BrFN2S: C, 52.90; H, 3.33; N, 7.71; S, 8.83; found: C, 52.92; H, 3.34; N,

380

7.70; S, 8.81; HR-MS (ESI+): m/z [M + H]

381

361.98890.

AC C

374

+

13

C NMR (101 MHz, DMSO-d6): δ

Calcd. for C16H12BrFN2S: 361.98886; found:

382 383

4.5.4. 5-Bromo-4-(4-bromophenyl)-N-(4-fluorophenyl)thiazol-2-amine (7d)

12

ACCEPTED MANUSCRIPT 384

White solid from EtOH (92% yield). m.p. 258.7-259.5°C; 1H NMR (400 MHz,

385

DMSO-d6): δ 10.32 (s, 1H), 8.88 (d, J = 8.7 Hz, 2H), 8.42 (d, J = 8.7 Hz, 2H), 7.53 (d, J =

386

8.1Hz, 2H), 6.80 (d, J = 8.1 Hz, 2H);

387

152.22, 138.30, 136.91, 134.14, 128.84, 123.01, 118.54, 116.33, 94.05; IR (KBr): 3438,

388

3068, 1625, 1598, 1511, 1236, 1001, 828, 644 cm-1. Anal. Calcd. for C15H9Br2FN2S: C,

389

42.08; H, 2.12; N, 6.54; S, 7.49; found: C, 42.10; H, 2.13; N, 6.57; S, 7.43; HR-MS (ESI+):

390

m/z [M + H] + Calcd. for C15H9Br2FN2S: 427.88168; found: 427.88168.

C NMR (101 MHz, DMSO-d6): δ 165.58, 158.06,

RI PT

13

391 392

4.5.5. N-(4-Fluorophenyl)-4-(naphthalen-2-yl)thiazol-2-amine (7e)

White solid from EtOH (91% yield). m.p. 171.5-175.3 °C; 1H NMR (400 MHz,

394

CDCl3): δ 8.35 (d, J = 1.3 Hz, 1H), 7.96 (dd, J = 6.1, 3.4 Hz, 1H), 7.89 (d, J = 8.6 Hz, 1H),

395

7.82 (dd, J = 6.0, 3.4 Hz, 1H), 7.75 (dd, J = 8.6, 1.9 Hz, 1H), 7.55 – 7.50 (m, 2H), 7.38 (dd, J

396

= 9.0, 4.5 Hz, 2H), 7.15 (dd, J = 8.9, 8.1 Hz, 2H), 6.78 (s, 1H), 5.27 (s, 1H); 13C NMR (101

397

MHz, CDCl3): δ 168.41, 133.84, 133.29, 129.48, 129.04, 127.75, 125.99 123.59, 122.66,

398

117.38, 117.15, 99.32; IR (KBr): 3251, 3049, 1625, 1503, 1231, 1158, 818, 750 cm-1. Anal.

399

Calcd. for C19H13FN2S: C, 71.23; H, 4.09; N, 8.74; S, 10.01; found: C, 71.24; H, 4.11; N,

400

8.75; S, 10.03. HR-MS (ESI+): m/z [M + H]

401

320.07839.

+

Calcd. for C19H13FN2S: 320.07835; found:

TE D

402 403

M AN U

SC

393

4.5.6. N-(4-Fluorophenyl)-4-(4-methoxyphenyl)thiazol-2-amine (7f) White solid from EtOH (87% yield). m.p. 231.5-234.3 °C; 1H NMR (400 MHz,

405

CDCl3): δ 11.54 (s, 1H), 7.72 (d, J = 8.9 Hz, 2H), 7.44 – 7.34 (m, 2H), 7.18 (dd, J = 9.1, 8.0

406

Hz, 2H), 7.01 (d, J = 8.9 Hz, 2H), 6.49 (s, 1H), 3.85 (s, 3H); 13C NMR (101 MHz, CDCl3): δ

407

164.08, 144.85, 142.28, 136.60, 129.41, 122.08, 121.99, 121.62, 120.49, 116.64, 116.41,

408

113.23, 17.46, 14.55; IR (KBr): 3212, 3015, 1619, 1505, 1292, 1255, 1022, 883 cm-1. Anal.

409

Calcd. for C16H13FN2OS: C, 63.98; H, 4.36; N, 9.39; S, 10.68; found: C, 63.99; H, 4.39; N,

410

9.37; S, 10.71. HR-MS (ESI+): m/z [M + H] + Calcd. for C16H13FN2OS: 300.07326; found:

411

300.07331.

AC C

EP

404

412 413

4.5.7. 4-(4-(4-Chlorophenyl)thiazol-2-ylamino)phenol (7g)

414

White solid from EtOH (87% yield). m.p. 196.3-198.3 °C; 1H NMR (400 MHz,

415

DMSO-d6): δ 9.93 (s, 1H), 9.15 (s, 1H), 7.90 (d, J = 8.7 Hz, 1H), 7.47 (d, J = 1.2 Hz, 1H),

416

7.45 (d, J = 1.4 Hz, 1H), 6.75 (d, J = 8.9 Hz, 1H); 13C NMR (101 MHz, DMSO-d6): δ 164.43,

417

152.42, 148.83, 133.54, 133.22, 131.85, 128.64, 127.36, 119.35, 115.52, 102.70; IR (KBr): 13

ACCEPTED MANUSCRIPT 418

3562, 3293, 3067, 1573, 1511, 1224, 672 cm-1. Anal. Calcd. for C15H11ClN2OS: C, 59.50; H,

419

3.66; N, 9.25; S, 10.59; found: C, 59.52; H, 3.68; N, 9.28; S, 10.62. HR-MS (ESI+): m/z [M

420

+ H] + Calcd. for C15H11ClN2OS: 302.02806; found: 302.02809.

421 422

4.5.8. 4-((4-(4-Methoxyphenyl)thiazol-2-yl)amino)phenol (7h) White solid from EtOH (88% yield). m.p. 126.3-128.3 °C; 1H NMR (400 MHz,

424

DMSO-d6): δ 9.93 (s, 1H), 9.15 (s, 1H), 7.90 (d, J = 8.7 Hz, 1H), 7.47 (d, J = 1.2 Hz, 1H),

425

7.45 (d, J = 1.4 Hz, 1H), 6.75 (d, J = 8.9 Hz, 1H), 3.85 (s, 3H );

426

DMSO-d6): δ 161.13, 151.39, 147.49, 132.45, 128.01, 125.22, 121.34, 117.15, 111.85,

427

104.90, 55.63; IR (KBr): 3562, 3293, 3067, 1573, 1511, 1224, 672 cm-1. Anal. Calcd. for

428

C15H11ClN2OS: C, 59.50; H, 3.66; N, 9.25; S, 10.59; found: C, 59.52; H, 3.68; N, 9.28; S,

429

10.62. HR-MS (ESI+): m/z [M + H]

430

302.02809.

RI PT

423

SC

C NMR (101 MHz,

Calcd. for C15H11ClN2OS: 302.02806; found:

M AN U

431 432

+

13

4.5.9. N-(4-Chloro-2-nitrophenyl)-4-(4-methoxyphenyl)thiazol-2-amine (7i) Yellow solid from EtOH (89% yield). m.p. 150.2-151.5 °C; 1H NMR (400 MHz,

434

DMSO-d6): δ 10.39 (s, 1H), 9.02 (d, J = 9.3 Hz, 1H), 8.37 (d, J = 2.6 Hz, 1H), 8.33 (dd, J =

435

9.3, 2.6 Hz, 1H), 7.93 (d, J = 8.7 Hz, 1H), 7.49 (s, 1H), 7.04 (d, J = 8.8 Hz, 1H), 3.85 (s, 1H);

436

13

437

124.15, 119.91, 117.73, 114.07, 105.16, 55.17; IR (KBr): 3390, 3102, 1590, 1503, 1335,

438

1280, 1176, 835, 739 cm-1. Anal. Calcd. for C16H12ClN3O3S: C, 53.11; H, 3.34; N, 11.61; S,

439

8.86; found: C, 53.14; H, 3.36; N, 11.68; S, 8.89. HR-MS (ESI+): m/z [M + H] + Calcd. for

440

C16H12ClN3O3S: 361.02879; found: 361.02883.

442 443

EP

C NMR (101 MHz, DMSO-d6): δ 161.41, 159.03, 149.71, 143.41, 140.19, 127.17, 125.09,

AC C

441

TE D

433

4.5.10. N-(4-Chloro-2-nitrophenyl)-4-(naphthalen-2-yl)thiazol-2-amine (7j) Yellow solid from EtOH (92% yield). m.p. 223.2-224.4 °C; 1H NMR (400 MHz,

444

DMSO-d6): δ 10.40 (s, 1H), 9.08 (d, J = 9.3 Hz, 1H), 8.48 (s, 1H), 8.34 (dd, J = 9.3, 2.4 Hz,

445

1H), 8.30 (d, J = 2.4 Hz, 1H), 8.07 (d, J = 8.6 Hz, 1H), 8.03 (d, J = 7.7 Hz, 1H), 7.95 (d, J =

446

8.6 Hz, 1H), 7.91 (d, J = 7.7 Hz, 1H), 7.73 (s, 1H), 7.56 – 7.46 (m, 1H); 13C NMR (101 MHz,

447

DMSO-d6): δ 161.58, 149.71, 143.28, 140.19, 133.14, 132.50, 128.25, 128.15, 127.51,

448

126.35, 126.05, 124.97, 124.41, 124.19, 123.90, 107. 86; IR (KBr): 3377, 3108, 1589, 1544,

449

1496, 1324, 1281, 1121, 892, 746 cm-1. Anal. Calcd. for C19H12ClN3O2S: C, 59.76; H, 3.17;

450

N, 11.00; S, 8.40; found: C, 59.79; H, 3.19; N, 11.04; S, 8.43. HR-MS (ESI+): m/z [M + H] +

451

Calcd. for C19H12ClN3O2S: 381.03388; found: 381.03393. 14

ACCEPTED MANUSCRIPT 452 453

4.5.11. N-(4-Fluorophenyl)-4-(pyridin-3-yl)thiazol-2-amine (7k)

454

Antique white solid from EtOH (91% yield). m.p. 181.5-188.1 °C; 1H NMR (400

455

MHz, CDCl3 + DMSO-d6): δ 10.19 (s, 1H), 9.60 (s, 1H), 9.10 (d, J = 1.6 Hz, 1H), 8.46 (dd, J

456

= 4.7, 1.3 Hz, 1H), 8.30 – 8.09 (m, 1H), 7.72 (dd, J = 9.1, 4.8 Hz, 2H), 7.36 (dd, J = 7.9, 4.8

457

Hz, 1H), 7.05 (t, J = 8.8 Hz, 2H);

458

147.85, 147.07, 137.35, 132.67, 130.08, 123.28, 118.26, 115.15, 114.93, 103.69; IR (KBr):

459

3443, 3114, 1619, 1576, 1508, 1402, 1225, 1021, 819, 710 cm-1. Anal. Calcd. for

460

C14H10FN3S: C, 61.98; H, 3.72; N, 15.49; S, 11.82; found: C, 61.99; H, 3.75; N, 15.54; S,

461

11.85. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H10FN3S: 271.05795; found: 271.05797.

C NMR (101 MHz, CDCl3 + DMSO-d6): δ 163.58,

SC

RI PT

13

462 463

4.5.12. N-(4-Fluorophenyl)-4-(pyridin-4-yl)thiazol-2-amine (7l)

Yellow solid from EtOH (88% yield). m.p. 283.5-284.3 °C; 1H NMR (400 MHz,

465

DMSO-d6): δ 10.72 (s, 1H), 8.91 (d, J = 6.7 Hz, 2H), 8.45 (d, J = 6.7 Hz, 2H), 8.33 (s, 1H),

466

7.79 (dd, J = 8.9, 4.8 Hz, 2H), 7.18 (t, J = 8.8 Hz, 2H); 13C NMR (101 MHz, DMSO-d6): δ

467

163.93, 156.00, 148.73, 145.25, 142.12, 137.13, 122.31, 118.82, 115.72, 115.50; IR (KBr):

468

3238, 3204, 3048, 1630, 1545, 1501, 1410, 1214, 826, 770 cm-1. Anal. Calcd. for

469

C14H10FN3S, C, 61.98; H, 3.72; N, 15.49; S, 11.82; found: C, 61.99; H, 3.75; N, 15.53; S,

470

11.85. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H10FN3S: 271.05795; found: 271.05798.

471

4.5.13. N-(4-Bromophenyl)-4-(pyridin-3-yl)thiazol-2-amine (7m)

TE D

M AN U

464

Yellow solid from EtOH (91% yield). m.p. 256.2-256.4 °C; 1H NMR (400 MHz,

473

DMSO-d6): δ 10.67 (s, 1H), 9.36 (d, J = 7.2 Hz, 1H), 9.06 – 8.88 (m, 1H), 8.86 – 8.74 (m,

474

1H), 8.06 (s, 1H), 7.91 (d, J = 10.4 Hz, 1H), 7.82 – 7.75 (m, 1H), 7.39 (d, J = 8.8 Hz, 1H);

475

13

476

109.09. IR (KBr): 3227, 3174, 3059, 1600, 1559, 1486, 1246, 820, 678 cm-1. Anal. Calcd. for

477

C14H10BrN3S: C, 50.61; H, 3.03; N, 12.65; S, 9.65; found: C, 50.64; H, 3.04; N, 12.68; S,

478

9.64. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H10BrN3S, 330.97788; found: 330.97792.

AC C

EP

472

C NMR (101 MHz, DMSO-d6): δ 163.62, 140.86, 139.63, 132.82, 128.77, 126.97, 118.60,

479 480

4.5.14. N-(4-Bromophenyl)-4-(pyridin-4-yl)thiazol-2-amine (7n)

481

Yellow solid from EtOH (94% yield). m.p. 281-282.5 °C; 1H NMR (400 MHz,

482

DMSO-d6): δ 10.82 (s, 1H), 8.92 (d, J = 6.8 Hz, 2H), 8.47 (d, J = 6.9 Hz, 2H), 8.37 (s, 1H),

483

7.80 (d, J = 9.0 Hz, 2H), 7.39 (d, J = 8.7 Hz, 2H); 13C NMR (101 MHz, DMSO-d6): δ 163.57,

15

ACCEPTED MANUSCRIPT 484

142.23, 128.88, 122.30, 118.71, 116.08; IR (KBr): 3221, 3173, 3043, 2717, 1631, 1544,

485

1488, 1310, 820, 667 cm-1. Anal. Calcd. for C14H10BrN3S: C, 50.61; H, 3.03; N, 12.65; S,

486

9.65; found: C, 50.62; H, 3.06; N, 12.68; S, 9.67. HR-MS (ESI+): m/z [M + H] + Calcd. for

487

C14H10BrN3S: 330.97788; found: 330.97793.

488

4.5.15. 4-(4-(Pyridin-3-yl)thiazol-2-ylamino)phenol (7o)

RI PT

489

Brown solid from EtOH (95% yield). m.p. 256.2-257.8 °C; 1H NMR (400 MHz,

491

CDCl3 + DMSO-d6): δ 9.66 (s, 1H), 9.05 (s, 1H), 8.84 (s, 1H), 8.43 (s, 1H), 8.13 (d, J = 7.7

492

Hz, 1H), 7.41 (d, J = 8.0 Hz, 2H), 7.33 (d, J = 4.3 Hz, 1H), 6.99 (s, 1H), 6.71 (d, J = 8.0 Hz,

493

2H); 13C NMR (101 MHz, CDCl3 + DMSO-d6): δ 164.92, 152.27, 147.39, 147.04, 146.52,

494

132.87, 119.29, 115.21, 102.35; IR (KBr): 3229, 3125, 1606, 1568, 1410, 1241, 831, 722 cm-

495

1

496

4.14; N, 15.63; S, 11.94. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H11N3OS: 269.06228;

497

found: 269.06231.

SC

490

M AN U

. Anal. Calcd. for C14H11N3OS: C, 62.43; H, 4.12; N, 15.60; S, 11.91; found: C, 62.45; H,

498 499

4.5.16. 4-(4-(Pyridin-4-yl)thiazol-2-ylamino)phenol (7p)

Dark red solid from EtOH (91% yield). m.p. 267.2-268.0 °C; 1H NMR (400 MHz,

501

DMSO-d6): δ 10.32 (s, 3H), 9.07 (m, 2H), 8.87 (d, J = 6.7 Hz, 6H), 8.41 (d, J = 6.8 Hz, 6H),

502

8.22 (s, 3H), 7.52 (d, J = 8.8 Hz, 6H), 6.79 (d, J = 8.8 Hz, 6H); 13C NMR (101 MHz, DMSO-

503

d6): δ 164.85, 152.80, 148.96, 145.36, 142.00, 132.71, 122.30, 119.61, 115.63, 115.02; IR

504

(KBr): 3468, 3215, 3025, 1636, 1561, 1433, 1259, 828, 747 cm-1. Anal. Calcd. for

505

C14H11N3OS, C, 62.43; H, 4.12; N, 15.60; S, 11.91; found: C, 62.45; H, 4.16; N, 15.63; S,

506

11.94. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H11N3OS: 269.06228; found: 269.0623.

EP

TE D

500

508 509

AC C

507

4.5.17. N-(4-Chloro-2-nitrophenyl)-4-(pyridin-2-yl)thiazol-2-amine (7q) Yellow solid from EtOH (92% yield). m.p. 311.6-312.5 °C; 1H NMR (400 MHz,

510

DMSO-d6): δ 11.91 (s, 1H), 8.60 (d, J = 7.8, 1H), 8.22 (d, J = 7.8, 1H), 8.05 (d, J = 7.8, 1H),

511

7.86 (t, J = 7.3, 1H), 7. 63 (d, J = 7.8, 1H), 7.42 – 7.37 (t, J = 7.3, 1H), 7.19 (s, 1H), 6.83 (d, J

512

= 7.7, 1H);

513

137.47, 135.24, 132.45, 127.45, 122.72, 121.60, 117.99, 105.18; IR (KBr): 3205, 3085, 1597,

514

1544, 1494, 1335, 1275, 756 cm-1. Anal. Calcd. for C14H9ClN4O2S: C, 50.53; H, 2.73; N,

515

16.84; S, 9.64; found: C, 50.55; H, 2.74; N, 16.86; S, 9.65. HR-MS (ESI+): m/z [M + H]

516

Calcd. for C14H9ClN4O2S: 332.01347; found: 332.01349.

13

C NMR (101 MHz, DMSO-d6): δ 161.41, 154.44, 149.98, 143.31, 141.64,

517 16

+

ACCEPTED MANUSCRIPT 518

4.5.18. N-(4-Chloro-2-nitrophenyl)-4-(pyridin-3-yl)thiazol-2-amine (7r)

519

Yellow solid from EtOH (88% yield). m.p.342.3-343.7 °C; 1H NMR (400 MHz,

520

DMSO-d6): δ 10.52 (s, 1H), 9.42 (s, 1H), 9.01 (d, J = 8.1 Hz, 1H), 8.96 (d, J = 9.3 Hz, 1H),

521

8.85 (d, J = 5.1 Hz, 1H), 8.28 (d, J = 2.2 Hz, 1H), 8.22 (dd, J = 9.3, 2.3 Hz, 1H), 8.14 (s, 1H),

522

8.08 (dd, J = 7.9, 5.8 Hz, 1H);

523

140.88, 140.31, 139.57, 132.33, 126.82, 124.75, 123.80, 120.11, 118.22, 112.47; IR (KBr):

524

3212, 3060, 1589, 1549, 1498, 1326, 1282, 1121, 816, 678 cm-1. Anal. Calcd. for

525

C14H9ClN4O2S: C, 50.53; H, 2.73; N, 16.84; S, 9.64; found: C, 50.56; H, 2.72; N, 16.85; S,

526

9.67. HR-MS (ESI+): m/z [M + H]

527

332.01352.

13

Calcd. for C14H9ClN4O2S: 332.01347; found:

SC

+

RI PT

C NMR (101 MHz, DMSO-d6): δ 162.45, 143.42, 142.62,

528 529

4.5.19. N-(4-Chloro-2-nitrophenyl)-4-(pyridin-4-yl)thiazol-2-amine (7s)

Yellow solid from EtOH (89% yield). m.p. 332.8 – 333.4 °C; 1H NMR (400 MHz,

531

DMSO-d6): δ 10.64 (s, 4H), 8.98 (d, J = 9.3 Hz, 6H), 8.91 (d, J = 5.4 Hz, 6H), 8.52 (s, 6H),

532

8.49 (d, J = 5.5 Hz, 5H), 8.36 (s, 3H), 8.28 (d, J = 8.9 Hz, 2H); 13C NMR (101 MHz, DMSO-

533

d6): δ 162.82, 147.85, 144.93, 142.77, 140.82, 129.29, 125.12, 123.98, 122.26, 120.60,

534

118.56; IR (KBr): 3231, 3046, 1632, 1602, 1548, 1502, 1324, 1120, 825, 738 cm-1. Anal.

535

Calcd. for C14H9ClN4O2S: C, 50.53; H, 2.73; N, 16.84; S, 9.64; found: C, 50.54; H, 2.75; N,

536

16.86; S, 9.65. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H9ClN4O2S: 332.01347; found:

537

332.01353.

TE D

M AN U

530

538

4.5.20. 4-(4-Methoxyphenyl)-N-(pyridin-2-yl)thiazol-2-amine (8a)

EP

539

Light yellow solid from EtOH (87% yield). m.p. 261.5-262.3 °C; 1H NMR (400 MHz,

541

DMSO-d6): δ 11.91 (s, 1H), 8.60 (d, J = 7.8 Hz, 1H), 8.05 (d, J = 7.8 Hz, 1H), 7.86 (t, J = 7.8

542

Hz, 1H), 7.65 (s, 1H) 7.52 (d, J = 8.2 Hz, 2H), 7.40 (t, J = 7.8 Hz, 1H), 7.07 (d, J = 8.2 Hz,

543

2H), 2.98 (s, 3H);

544

138.30, 128.01, 124.39, 117.42, 114.36, 108.25, 103.23, 55.92; IR (KBr): 3231, 3077, 1618,

545

1559, 1474, 1429, 1344, 1238, 771, 671 cm-1. Anal. Calcd. for C15H13N3OS: C, 63.58; H,

546

4.62; N, 14.83; S, 11.32; found: C, 63.55; H, 4.65; N, 14.86; S, 11.36. HR-MS (ESI+): m/z

547

[M + H] + Calcd. for C15H13N3OS: 283.07793; found: 283.07795.

AC C

540

13

C NMR (101 MHz, DMSO-d6): δ 162.24, 154.73, 150.56, 147.77,

548 549

4.5.21. 4-(4-Bromophenyl)-N-(pyridin-2-yl)thiazol-2-amine (8b)

550

Light yellow solid from EtOH (92% yield). m.p. 281.5-282.1 °C; 1H NMR (400 MHz,

551

DMSO-d6): δ 12.49 (s, 1H), 8.44 (s, 1H), 7.96 (d, J = 3.8 Hz, 1H), 7.80 (d, J = 7.8 Hz, 2H), 17

ACCEPTED MANUSCRIPT 13

7.51 (d, J = 6.6 Hz, 2H), 7.42 (s, 1H), 7.36 (s, 1H), 7.14 (s, 1H);

553

DMSO-d6): δ 161.41, 155.84, 149.98, 148.04, 138.30, 132.73, 131.35, 128.01, 123.27,

554

117.99, 107.69, 104.61, 40.15, 39.94, 39.74, 39.53, 39.32, 39.11, 38.90; IR (KBr): 3231,

555

3077, 1618, 1559, 1474, 1429, 1344, 1238, 771, 671 cm-1. Anal. Calcd. for C14H10BrN3S: C,

556

50.61; H, 3.03; N, 12.65; S, 9.65; found: C, 50.64; H, 3.05; N, 12.67; S, 9.68 HR-MS (ESI+):

557

m/z [M + H] + Calcd. for C14H10BrN3S: 330.97788; found: 330.97788.

558 559

C NMR (101 MHz,

RI PT

552

4.5.22. 4-(4-Bromophenyl)-5-methyl-N-(pyridin-2-yl)thiazol-2-amine (8c)

White solid from EtOH (90% yield). m.p. 277.5-278.2 °C; 1H NMR (400 MHz,

561

DMSO-d6): δ 11.85 (s, 1H), 8.35 (dd, J = 5.4, 1.0 Hz, 1H), 7.90 – 7.85 (m, 1H), 7.64 (d, J =

562

2.9 Hz, 2H), 7.21 (d, J = 8.5 Hz, 1H), 7.09 – 7.04 (m, 1H), 2.45 (s, 1H); 13C NMR (101 MHz,

563

DMSO-d6): δ 161.52, 156.39, 150.39, 144.03, 141.70, 133.22, 131.37, 130.22, 120.72,

564

120.46, 116.65, 112.07, 11.82; IR (KBr): 3231, 3077, 1618, 1559, 1474, 1429, 1344, 1238,

565

771, 671 cm-1. Anal. Calcd. for C15H12BrN3S: C, 52.03; H, 3.49; N, 12.14; S, 9.26; found: C,

566

52.06; H, 3.51; N, 12.17; S, 9.28. HR-MS (ESI+): m/z [M + H] + Calcd. for C15H12BrN3S:

567

344.99353; found: 344.99356.

568 569

M AN U

SC

560

4.5.23. 5-Bromo-4-(4-bromophenyl)-N-(pyridin-2-yl)thiazol-2-amine (8d) White solid from EtOH (86% yield). m.p. 230.1-232 °C; 1H NMR (400 MHz, DMSO-

571

d6): δ 11.89 (s, 1H), 8.38 (d, J = 4.8 Hz, 1H), 7.90 (d, J = 8.3 Hz, 2H), 7.62 (s, 1H), 7.60 (d, J

572

= 2.7 Hz, 1H), 7.23 (d, J = 8.4 Hz, 1H), 7.09 – 7.04 (m, 1H); 13C NMR (101 MHz, DMSO-

573

d6): δ 159.96, 150.63, 147.58, 144.18, 140.05, 133.38, 131.61, 127.88, 120.83, 116.54,

574

112.03, 107.50; IR (KBr): 3474, 3070, 1649, 1615, 1525, 1452, 1198, 832, 767, 668 cm-1.

575

Anal. Calcd. for C14H9Br2N3S: C, 40.90; H, 2.21; N, 10.22; S, 7.80; found: C, 40.93; H, 2.23;

576

N, 10.25; S, 7.84. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H9Br2N3S: 408.88839; found:

577

408.88842.

579

EP

AC C

578

TE D

570

4.5.24. 4-(3,4-Dichlorophenyl)-N-(3-methylpyridin-2-yl)thiazol-2-amine (8e)

580

White solid from EtOH (88% yield). m.p. 276.2-278.1 °C; 1H NMR (400 MHz,

581

DMSO-d6): δ 11.21 (s, 1H), 8.06 (d, J = 8.9 Hz, 1H), 7.95 (s, 1H), 7.86 (d, J = 5.2 Hz, 1H),

582

7.62 (s, 1H), 7.49 (d, J = 7.5 Hz, 1H), 7.05 (t, J = 7.5 Hz, 1H), 6.91 (d, J = 7.8 Hz, 1H), 2.46

583

(s, 3H); 13C NMR (101 MHz, DMSO-d6): δ 163.08, 160.02, 151.66, 146.10, 138.30, 134.40,

584

131.90, 131.84, 129.39, 129.11, 125.78, 117.71, 113.52, 106.02; IR (KBr): 3421, 3083, 1637,

585

1583, 1489, 1258, 1191, 783, 738 cm-1. Anal. Calcd. for C15H11Cl2N3S: C, 53.58; H, 3.30; N, 18

ACCEPTED MANUSCRIPT 586

12.50; S, 9.54; found: C, 53.62; H, 3.32; N, 12.55; S, 9.56. HR-MS (ESI+): m/z [M + H]

587

Calcd. for C15H11Cl2N3S: 335.00507; found: 335.00509.

+

588 589

4.5.25. 4-(4-Bromophenyl)-5-methyl-N-(3-methylpyridin-2-yl)thiazol-2-amine (8f) Light Yellow solid from EtOH (94% yield). m.p. 243.1-244.5 °C; 1H NMR (400

591

MHz, DMSO-d6): δ 11.73 (s, 1H), 7.77 (t, J = 7.3 Hz, 1H), 7.69 (dd, J = 16.6, 8.5 Hz, 1H),

592

7.02 (d, J = 8.1 Hz, 1H), 6.95 (d, J = 7.2 Hz, 1H), 2.55 (s, 1H); 13C NMR (101 MHz, DMSO-

593

d6): δ 162.80, 157.78, 147.77, 145.25, 136.64, 133.29, 131.62, 128.56, 123.82, 117.42,

594

115.76, 113.52; IR (KBr): 3444, 3073, 1653, 1586, 1455, 1202, 1167, 997, 806, 667 cm-1.

595

Anal. Calcd. for C16H14BrN3S: C, 53.34; H, 3.92; N, 11.66; S, 8.90; found: C, 53.36; H, 3.95;

596

N, 11.69; S, 8.94. HR-MS (ESI+): m/z [M + H] + Calcd. for C16H14BrN3S: 359.00918; found:

597

359.00921.

SC

RI PT

590

599

M AN U

598

4.5.26. 5-Bromo-4-(4-bromophenyl)-N-(3-methylpyridin-2-yl)thiazol-2-amine (8g) White solid from EtOH (87% yield). m.p. 286.9-288.1 °C; 1H NMR (400 MHz,

601

DMSO-d6): δ 11.34 (s, 1H), 8.01 (d, J = 8.9 Hz, 1H), 7.76 (d, J = 8.6 Hz, 2H), 7.56 (d, J =

602

8.6 Hz, 2H), 7.26 (d, J = 10.9 Hz, 1H), 6.62 (t, J = 8.9 Hz, 1H), 2.30 (s, 1H); 13C NMR (101

603

MHz, DMSO-d6) δ 164.05, 161.00, 152.38, 144.08, 136.59, 132.15, 132.03, 128.93, 123.85,

604

118.76, 112.83, 93.34, 16.79; IR (KBr): 3439, 3072, 1653, 1588, 1453, 1211, 1165, 985, 811,

605

655 cm-1. Anal. Calcd. for C15H11Br2N3S: C, 42.38; H, 2.61; N, 9.88; S, 7.54; found: C,

606

42.39; H, 2.66; N, 9.91; S, 7.56. HR-MS (ESI+): m/z [M + H]

607

424.90200; found: 424.9023.

608

610

Calcd. for C15H11Br2N3S:

4.5.27. 4-(3,4-Dichlorophenyl)-N-(6-methylpyridin-2-yl)thiazol-2-amine (8h) White solid from EtOH (94% yield). m.p. 281.3-282 °C; 1H NMR (400 MHz, DMSO-

AC C

609

+

EP

TE D

600

611

d6): δ 11.91 (s, 1H), 8.60 (d, J = 3.8 Hz, 1H), 8.05 (d, J = 7.8 Hz, 1H), 7.86 (t, J = 7.3 Hz,

612

1H), 7.57 (s, 1H), 7.46 – 7.32 (m, 1H), 7.14 (d, J = 7.1 Hz, 1H), 6.96 (d, J = 2.7 Hz, 1H),

613

2.52 (S, 1H);

614

133.74, 132.05, 129.50, 127.52, 126.67, 120.18, 107.19, 104.92, 21.58; IR (KBr): 3385,

615

3075, 2857, 1631, 1585, 1452,1392, 1314, 1260, 1188,1071, 1003, 910, 832, 784 cm-1. Anal.

616

Calcd. for C15H11Cl2N3S: C, 53.58; H, 3.30; N, 12.50; S, 9.54; found: C, 53.60; H, 3.34; N,

617

12.53; S, 9.55. HR-MS (ESI+): m/z [M + H] + Calcd. for C15H11Cl2N3S: 335.00507; found:

618

335.00510.

13

C NMR (101 MHz, DMSO-d6) δ 160.86, 155.20, 153.22, 150.11, 138.26,

619 19

ACCEPTED MANUSCRIPT 620

4.5.28. 4-(4-Bromophenyl)-5-methyl-N-(6-methylpyridin-2-yl)thiazol-2-amine (8i)

621

White solid from EtOH (93% yield). m.p. 282.1-282.9 °C; 1H NMR (400 MHz,

622

DMSO-d6): δ 11.91 (s, 1H), 8.60 (d, J = 3.8 Hz, 1H), 7.87 (d, J = 7.3 Hz, 1H), 7.40 (t, J =

623

7.3, 1H), 6.92 (d, J = 6.3 Hz, 1H), 6.59 (d, J = 8.4 Hz, 1H), 2.50 (s, 4H), 2.40 (s, 3H);

624

NMR (101 MHz, DMSO-d6) δ 162.26, 158.31, 153.22, 148.99, 137.41, 132.31, 131.19,

625

128.38, 123.01, 119.89, 117.63, 103.52, 24.41, 13.97; IR (KBr): 3519, 3074, 1658, 1584,

626

1216, 1168, 987, 821, 643 cm-1. Anal. Calcd. for C16H14BrN3S: C, 53.34; H, 3.92; N, 11.66;

627

S, 8.90; found: C, 53.36; H, 3.89; N, 11.69; S, 8.92. HR-MS (ESI+): m/z [M + H] + Calcd. for

628

C16H14BrN3S: 359.00918; found: 359.00920.

RI PT

C

SC

629 630

13

4.5.29. 4-(4-Methoxyphenyl)-N-(6-methylpyridin-2-yl)thiazol-2-amine (8j) Antique white solid from EtOH (89% yield). m.p. 221.5-222.5 °C; 1H NMR (400

632

MHz, CDCl3 + DMSO-d6): δ 7.89 (s, 1H), 7.68 (d, J = 8.7 Hz, 2H), 7.29 (d, J = 7.1 Hz, 1H),

633

7.18 (s, 1H), 7.05 (d, J = 7.3 Hz, 1H), 6.92 (d, J = 8.8 Hz, 2H), 4.39 (s, 1H), 3.77 (s, 4H),

634

2.64 (s, 3H);

635

136.96, 129.23, 126.17, 116.70, 113.28, 109.67, 103.86, 54.20, 22.47; IR (KBr): 3518, 3068,

636

1644, 1580, 1454, 1256, 1172, 1029, 799 cm-1. Anal. Calcd. for C16H15N3OS, C, 64.62; H,

637

5.08; N, 14.13; S, 10.78; found: C, 64.65; H, 5.10; N, 14.17; S, 10.81. HR-MS (ESI+): m/z

638

[M + H] + Calcd. for C16H15N3OS: 297.09358; found: 297.09361.

640

13

C NMR (101 MHz, CDCl3 + DMSO-d6): δ 163.65, 159.06, 157.67, 150.31,

TE D

639

M AN U

631

4.5.30. N-(6-Methylpyridin-2-yl)-4-(naphthalen-1-yl)thiazol-2-amine (8k) White Solid from EtOH (94% yield). m.p. 227.5-228.4 °C; 1H NMR (400 MHz,

642

DMSO-d6): δ 11.68 (s, 1H), 8.42 (s, 1H), 8.04 (dd, J = 8.6, 1.7 Hz, 1H), 7.99 – 7.86 (m, 2H),

643

7.69 – 7.45 (m, 2H), 7.02 – 6.77 (m, 1H), 2.47 (s, 1H).

644

158.36, 155.05, 150.37, 145.25, 138.56, 128.23, 127.73, 127.55, 127.07, 126.52, 125.63,

645

115.48, 107.56, 94.97, 23.16; IR (KBr): 3057, 3028, 2703, 1649, 1590, 1457, 1170, 885, 784,

646

746 cm-1. Anal. Calcd. for C19H15N3S: C, 71.90; H, 4.76; N, 13.24; S, 10.1; found: 1 C,

647

71.92; H, 4.79; N, 13.28; S, 10.12. HR-MS (ESI+): m/z [M + H]

648

317.09867; found: 317.09872.

13

C NMR (101 MHz, DMSO-d6): δ

AC C

EP

641

+

Calcd. for C19H15N3S:

649 650

4.5.31. N-(Pyridin-2-yl)-4-(pyridin-3-yl)thiazol-2-amine (8l)

651

Antique white solid from EtOH (94% yield). m.p. 235.6-237.6 °C; 1H NMR (400

652

MHz, DMSO-d6): δ 11.48 (s, 1H), 9.14 (s, 1H), 8.51 (d, J = 3.9 Hz, 1H), 8.32 (dd, J = 5.0,

653

1.0 Hz, 1H), 8.28 – 8.21 (m, 1H), 7.72 (ddd, J = 8.8, 7.3, 1.8 Hz, 1H), 7.61 (s, 1H), 7.11 (d, J 20

ACCEPTED MANUSCRIPT 13

C NMR (101 MHz, DMSO-d6): δ 160.01,

654

= 8.3 Hz, 1H), 6.94 (dd, J = 6.4, 5.2 Hz, 1H);

655

151.70, 148.19, 146.78, 146.43, 137.97, 132.79, 123.79, 116.12, 110.86, 107.53; IR (KBr):

656

3241, 3097, 2913, 1621, 1547, 1483, 1404, 1311, 765, 715 cm-1. Anal. Calcd. for C13H10N4S:

657

C, 61.40; H, 3.96; N, 22.03; S, 12.61; found: C, 61.43; H, 3.99; N, 22.07; S, 12.62. HR-MS

658

(ESI+): m/z [M + H] + Calcd. for C13H10N4S: 254.06262; found: 254.06266.

660

RI PT

659

4.5.32. N-(3-Methylpyridin-2-yl)-4-(pyridin-3-yl)thiazol-2-amine (8m)

661

Yellow solid from EtOH (89% yield). m.p. 160.2-161.4 °C; 1H NMR (400 MHz,

662

DMSO-d6): δ 10.54 (s, 1H), 9.16 (d, J = 1.7 Hz, 1H), 8.49 (dd, J = 4.7, 1.5 Hz, 1H), 8.28 –

663

8.23 (m, 1H), 8.18 (dd, J = 4.9, 1.1 Hz, 1H), 7.60 (s, 1H), 7.57 – 7.52 (m, 1H), 7.43 (ddd, J =

664

8.0, 4.8, 0.7 Hz, 1H), 6.90 (dd, J = 7.2, 5.0 Hz, 1H), 2.35 (s, 3H);

665

CDCl3): δ 160.56, 149.64, 148.56, 147.57, 146.41, 144.29, 138.31, 133.19, 130.79, 123.55,

666

117.84, 116.66, 107.51, 16.67; IR (KBr): 3219, 3102, 2921, 1591, 1463, 1189, 811, 781, 731

667

cm-1.. Anal. Calcd. for C14H12N4S C, 62.66; H, 4.51; N, 20.88; S, 11.9; found: 5 C, 62.68; H,

668

4.53; N, 20.94; S, 11.98. HR-MS (ESI+): m/z [M + H]

669

found: 268.07831.

SC

C NMR (101 MHz,

M AN U

670 671

13

+

Calcd. for C14H12N4S: 268.07827;

4.5.33. N-(6-Methylpyridin-2-yl)-4-(pyridin-4-yl)thiazol-2-amine (8n) Yellow solid from EtOH (94% yield). m.p. 294.5-295.4 °C; 1H NMR (400 MHz,

673

DMSO-d6): δ 11.62 (s, 1H), 8.93 (d, J = 6.9 Hz, 1H), 8.42 (d, J = 6.8 Hz, 1H), 8.40 (s, 1H),

674

7.67 – 7.61 (m, 1H), 6.93 (d, J = 8.2 Hz, 1H), 6.84 (d, J = 7.3 Hz, 1H), 2.48 (s, 1H);

675

NMR (101 MHz, DMSO-d6): δ 166.66, 160.71, 155.13, 150.52, 143.66, 142.25, 138.58,

676

122.01, 118.20, 115.59, 107.68, 23.24; IR (KBr): 3211, 3187, 3093, 2729, 1629, 1604, 1541,

677

1463, 1323, 1199, 828, 777 cm-1. Anal. Calcd. for C14H12N4S: C, 62.66; H, 4.51; N, 20.88; S,

678

11.95; found: C, 62.67; H, 4.55; N, 20.94; S, 11.98. HR-MS (ESI+): m/z [M + H] + Calcd. for

679

C14H12N4S: 268.07827; found: 268.07829.

681

13

EP

C

AC C

680

TE D

672

4.5.34. N-(3-Methylpyridin-2-yl)-4-(pyridin-4-yl)thiazol-2-amine (8o)

682

Yellow solid from EtOH (88% yield). m.p. 314.5-315.9 °C; 1H NMR (400 MHz,

683

DMSO-d6): δ 11.15 (s, 1H), 8.98 (d, J = 6.6 Hz, 2H), 8.55 (d, J = 6.6 Hz, 2H), 8.51 (s, 1H),

684

8.28 (d, J = 6.7 Hz, 1H), 7.73 (d, J = 6.7 Hz, 1H), 7.10 – 7.01 (t, J = 6.7 Hz, 1H); 13C NMR

685

(101 MHz, DMSO-d6) δ 161.98, 157.74, 148.99, 145.60, 140.78, 137.69, 136.56, 123.85,

686

116.80, 113.12, 108.88, 40.15, 39.94, 39.74, 39.53, 39.32, 39.11, 38.90, 16.22; IR (KBr):

687

3472, 3390, 3079, 1635, 1581, 1530, 1499, 1458, 1373, 1256, 1190, 827, 790, 670 cm-1. 21

ACCEPTED MANUSCRIPT 688

Anal. Calcd. for C14H12N4S: C, 62.66; H, 4.51; N, 20.88; S, 11.95; found: C, 62.67; H, 4.55;

689

N, 20.94; S, 11.98. HR-MS (ESI+): m/z [M + H] + Calcd. for C14H12N4S: 268.07827; found:

690

268.07829.

691

4.6.

In vitro assay for evaluation of antimycobacterial activity

RI PT

692

The in vitro assay method previously reported by our group has been used for

694

evaluation of antimycobacterial assay. DMSO was used for the preparation of the test

695

compound stocks as well as for dilutions. The test compounds MICs against Mtb were

696

analysed in 7H9 broth using a standard micro dilution method [61]. The test compounds

697

which were prepared with 1µl of serial twofold dilutions in DMSO were added in to the 384

698

well plates with the final concentrations ranging from 100 mM - 0.19 mM. Two sets of

699

control wells were prepared: a media control with only medium (Middlebrook 7H9 medium

700

supplemented with 0.2% glycerol, 0.05% Tween 80 (Sigma), and 10% albumin dextrose

701

catalase (Difco Laboratories, Detroit, Mich.)) and a culture control with a bacterial

702

suspension but no compound. 40µl (3-7 x 105 CFU/ml) of the bacterial culture was added to

703

all the wells except the media control wells. The plates were packed in gas permeable

704

polythene bags and incubated at 37°C for 5 days. Following this incubation period, 8 µl of a

705

freshly prepared 1:1 mixture of Resazurin (0.02% in water) and 10% Tween 80 was added to

706

all the wells. The plates were re-incubated for an additional 24 h at 37°C and the colour

707

conversion of all wells recorded. A blue colour in the well indicates no growth and a pink

708

colour indicates growth. Absorbance at 575 nm and 610 nm was monitored and the ratio

709

(A575/A610) calculated. The least concentration which yielded 80% inhibition was considered

710

as MIC; the media control is considered 100% inhibition and the culture control as 0%

711

inhibition. Isoniazid was used as a reference drug for the assay.

712

4. 7.

M AN U

TE D

EP

AC C

713

SC

693

Molecular docking studies

The docking simulations were performed using Auto Dock 4.2 [62]. The recently

714

solved X-ray crystal structure of KasA of Mtb (PDB code: 2WGD) in complex with TLM has

715

been retrieved from the RCSB Protein Data Bank and all heteroatoms were removed. The

716

2WGD was setup for a standard protocol for docking. Lamarckian Genetic Algorithm was

717

used to carry out the simulation. Columbic electrostatic potential, Vander Waals interaction

718

represented as a Lennard-Jones12–6 dispersion/repulsion term and hydrogen bonding 22

ACCEPTED MANUSCRIPT represented as a directional 12–10 term were the three parameters taken into account for

720

evaluating binding energy in the docking step. The most favourable free energy of binding

721

were attained by considering the docking orientations lying within the range of 2.0 Å in the

722

root-mean square deviation (rmsd) tolerance and clustering the each other to get the result.

723

The post-docking energy minimization on Discovery Studio 2.5 was performed on top-posed

724

docking conformations obtained.

725

Acknowledgements

RI PT

719

We are grateful to AstraZeneca India Pvt. Ltd., Bangalore, India for generation of the

726

MIC data against Mtb. The authors thank Council of Scientific and Industrial Research

728

(CSIR), New Delhi, India for financial support through a sponsored project (01 (2262)

729

/08/EMR-II). Mr. Parameshwar Makam thanks Pondicherry University for awarding

730

University Research Fellowship (URF).

731

Appendix A. Supplementary information

M AN U

SC

727

Supplementary information related to this article can be found at…

732 733

References

734

[1]

TE D

Tuberculosis: WHO global tuberculosis report 2013, in, World Health Organization, World Health Organization, Geneva, 2013.

735

[2]

H. Herzog, History of tuberculosis, Respiration 65 (1998) 5-15.

737

[3]

C. Lienhardt, S.V. Cook, M. Burgos, V.Y. Edwards, L. Rigouts, G. Anyo, K.S. Jae,

EP

736

A. Jindani, D.A. Enarson, A.J. Nunn, Efficacy and safety of a 4-drug fixed-dose

739

combination regimen compared with separate drugs for treatment of pulmonary

740

tuberculosis: The study c randomized controlled trial, JAMA 305 (2011) 1415-1423.

741

[4]

359 (2002) 2188-2194.

743

[5]

[6]

749

R. Osborne, First novel anti-tuberculosis drug in 40 years, Nat. Biotech. 31 (2013) 8991.

747 748

M.R. Ward, Notes on the 2004 to 2009 United States Food and Drug Administration approval of new molecular entities (NMEs), in: KEI Research Note, 2010, pp. 1-14.

745 746

P. Trouiller, P. Olliaro, E. Torreele, J. Orbinski, R. Laing, N. Ford, Drug development for neglected diseases: A deficient market and a public-health policy failure, Lancet

742

744

AC C

738

[7]

A. Zumla, P. Nahid, S.T. Cole, Advances in the development of new tuberculosis drugs and treatment regimens, Nat. Rev. Drug Discov. 12 (2013) 388-404. 23

ACCEPTED MANUSCRIPT 750

[8]

Microbiol. Rev. 24 (2011) 351-376.

751 752

[9]

Z.F. Udwadia, R.A. Amale, K.K. Ajbani, C. Rodrigues, Totally drug-resistant tuberculosis in India, Clin. Infect. Dis. 54 (2012) 579-581.

753 754

C.K. Kwan, J.D. Ernst, HIV and tuberculosis: A deadly human syndemic, Clin.

[10]

M. Zignol, M. S.Hosseini, A. Wright, C.L.V. Weezenbeek, P. Nunn, C.J. Watt, B.G. Williams, C. Dye, Global incidence of multidrug-resistant tuberculosis, J. Infect. Dis.

756

194 (2006) 479-485.

757

[11]

RI PT

755

N.R. Gandhi, A. Moll, A.W. Sturm, R. Pawinski, T. Govender, U. Lalloo, K. Zeller, J. Andrews, G. Friedland, Extensively drug-resistant tuberculosis as a cause of death in

759

patients co-infected with tuberculosis and HIV in a rural area of South Africa, Lancet

760

368 (2006) 1575-1580. [12]

S.E. Dorman, R.E. Chaisson, From magic bullets back to the magic mountain: The rise of extensively drug-resistant tuberculosis, Nat. Med. 13 (2007) 295-298.

762

M AN U

761

SC

758

763

[13]

C. Dye, Global epidemiology of tuberculosis, Lancet 367 (2006) 938-940.

764

[14]

C.U. Köser, J.M. Bryant, J. Becq, M.E. Török, M.J. Ellington, M.A. Marti-Renom,

765

A.J. Carmichael, J. Parkhill, G.P. Smith, S.J. Peacock, Whole-genome sequencing for

766

rapid susceptibility testing of M. tuberculosis, New Engl. J. Med. 369 (2013) 290-292. [15]

M.R. Farhat, B.J. Shapiro, K.J. Kieser, R. Sultana, K.R. Jacobson, T.C. Victor, R.M.

TE D

767

Warren, E.M. Streicher, A. Calver, A. Sloutsky, D. Kaur, J.E. Posey, B. Plikaytis,

769

M.R. Oggioni, J.L. Gardy, J.C. Johnston, M. Rodrigues, P.K.C. Tang, M. Kato-

770

Maeda, M.L. Borowsky, B. Muddukrishna, B.N. Kreiswirth, N. Kurepina, J. Galagan,

771

S. Gagneux, B. Birren, E.J. Rubin, E.S. Lander, P.C. Sabeti, M. Murray, Genomic

772

analysis identifies targets of convergent positive selection in drug-resistant

773

Mycobacterium tuberculosis, Nat. Genet. 45 (2013) 1183-1189. [16]

AC C

774

EP

768

H. Zhang, D. Li, L. Zhao, J. Fleming, N. Lin, T. Wang, Z. Liu, C. Li, N. Galwey, J.

Deng, Y. Zhou, Y. Zhu, Y. Gao, T. Wang, S. Wang, Y. Huang, M. Wang, Q. Zhong,

775

L. Zhou, T. Chen, J. Zhou, R. Yang, G. Zhu, H. Hang, J. Zhang, F. Li, K. Wan, J.

776

Wang, X.-E. Zhang, L. Bi, Genome sequencing of 161 Mycobacterium tuberculosis

777 778

isolates from China identifies genes and intergenic regions associated with drug

779

resistance, Nat. Genet. 45 (2013) 1255-1260.

780

[17]

S. Ananthan, E.R. Faaleolea, R.C. Goldman, J.V. Hobrath, C.D. Kwong, B.E.

781

Laughon, J.A. Maddry, A. Mehta, L. Rasmussen, R.C. Reynolds, J.A. Secrist, 3rd, N.

782

Shindo, D.N. Showe, M.I. Sosa, W.J. Suling, E.L. White, High-throughput screening

783

for inhibitors of Mycobacterium tuberculosis H37Rv, Tuberculosis 89 (2009) 334-353. 24

ACCEPTED MANUSCRIPT 784

[18]

R.C. Reynolds, S. Ananthan, E. Faaleolea, J.V. Hobrath, C.D. Kwong, C. Maddox, L.

785

Rasmussen, M.I. Sosa, E. Thammasuvimol, E.L. White, W. Zhang, J.A. Secrist Iii,

786

High throughput screening of a library based on kinase inhibitor scaffolds against

787

Mycobacterium tuberculosis H37Rv, Tuberculosis 92 (2012) 72-83. [19]

development, J. Med. Chem. 54 (2011) 6157-6165.

789 790

[20]

A. Koul, E. Arnoult, N. Lounis, J. Guillemont, K. Andries, The challenge of new drug discovery for tuberculosis, Nature 469 (2011) 483-490.

791 792

L.G. Dover, G.D. Coxon, Current status and research strategies in tuberculosis drug

[21]

RI PT

788

M. Pieroni, B. Wan, S. Cho, S.G. Franzblau, G. Costantino, Design, synthesis and investigation on the structure–activity relationships of N-substituted 2-aminothiazole

794

derivatives as antitubercular agents, Eur. J. Med. Chem. 72 (2014) 26-34.

795

[22]

SC

793

A. Meissner, H.I. Boshoff, M. Vasan, B.P. Duckworth, C.E. Barry Iii, C.C. Aldrich, Structure–activity relationships of 2-aminothiazoles effective against Mycobacterium

797

tuberculosis, Bioorg. Med. Chem. 21 (2013) 6385-6397.

798

[23]

M AN U

796

G. Pappenberger, T. Schulz-Gasch, E. Kusznir, F. Muller, M. Hennig, Structureassisted discovery of an aminothiazole derivative as a lead molecule for inhibition of

800

bacterial fatty-acid synthesis, Acta Crystallogr., Sect. D: Biol. Crystallogr. 63 (2007)

801

1208-1216.

802

[24]

TE D

799

S.R. Luckner, C.A. Machutta, P.J. Tonge, C. Kisker, Crystal structures of

803

Mycobacterium tuberculosis KasA show mode of action within cell wall biosynthesis

804

and its inhibition by thiolactomycin, Structure 17 (2009) 1004-1013. [25]

Q. Al-Balas, N.G. Anthony, B. Al-Jaidi, A. Alnimr, G. Abbott, A.K. Brown, R.C.

EP

805

Taylor, G.S. Besra, T.D. McHugh, S.H. Gillespie, B.F. Johnston, S.P. Mackay, G.D.

807

Coxon, Identification of 2-aminothiazole-4-carboxylate derivatives active against Mycobacterium tuberculosis H37Rv and the β-ketoacyl-ACP synthase MtfabH, PloS

808

one 4 (2009) e5617.

809 810

[26]

E.C. Dykhuizen, J.F. May, A. Tongpenyai, L.L. Kiessling, Inhibitors of UDP-

Galactopyranose mutase thwart mycobacterial growth, J. Am. Chem. Soc. 130 (2008)

811

6706-6707.

812 813

AC C

806

[27]

L.P.S. de Carvalho, G. Lin, X. Jiang, C. Nathan, Nitazoxanide kills replicating and

814

nonreplicating Mycobacterium tuberculosis and evades resistance, J. Med. Chem. 52

815

(2009) 5789-5792.

816 817

[28]

L.M. Fox, L.D. Saravolatz, Nitazoxanide: A new thiazolide antiparasitic agent, Clin. Infect. Dis. 40 (2005) 1173-1180. 25

ACCEPTED MANUSCRIPT 818

[29]

imidazo[2,1-b]thiazole derivatives, Eur. J. Med. Chem. 42 (2007) 320-326.

819 820

[30]

J. Araujo, C. Logothetis, Dasatinib: A potent SRC inhibitor in clinical development for the treatment of solid tumors, Cancer Treat. Rev. 36 (2010) 492-500.

821 822

E. Gürsoy, N.U. Güzeldemirci, Synthesis and primary cytotoxicity evaluation of new

[31]

W.C. Patt, H.W. Hamilton, M.D. Taylor, M.J. Ryan, D.G. Taylor, C.J.C. Connolly, A.M. Doherty, S.R. Klutchko, I. Sircar, Structure-activity relationships of a series of

824

2-amino-4-thiazole-containing renin inhibitors, J. Med. Chem. 35 (1992) 2562-2572.

825

[32]

RI PT

823

O. Kouatly, A. Geronikaki, C. Kamoutsis, D. Hadjipavlou-Litina, P. Eleftheriou,

Adamantane derivatives of thiazolyl-N-substituted amide, as possible non-steroidal

827

anti-inflammatory agents, Eur. J. Med. Chem. 44 (2009) 1198-1204.

828

[33]

SC

826

J.S. Carter, D.J. Rogier, M.J. Graneto, K. Seibert, C.M. Koboldt, Z. Yan, J.J. Talley, Design and synthesis of sulfonyl-substituted 4,5-diarylthiazoles as selective

830

cyclooxygenase-2 inhibitors, Bioorg. Med. Chem. Lett. 9 (1999) 1167-1170.

831

[34]

M AN U

829

N. Ergenç, G. Çapan, N.S. Günay, S. Özkirimli, M. Güngör, S. Özbey, E. Kendi,

832

Synthesis and hypnotic activity of new 4-thiazolidinone and 2-thioxo-4,5-

833

imidazolidinedione derivatives, Arch. Pharm. 332 (1999) 343-347.

834

[35]

J.C. Jaen, L.D. Wise, B.W. Caprathe, H. Tecle, S. Bergmeier, C.C. Humblet, T.G. Heffner, L.T. Meltzer, T.A. Pugsley, 4-(1,2,5,6-Tetrahydro-1-alkyl-3-pyridinyl)-2-

836

thiazolamines: A novel class of compounds with central dopamine agonist properties,

837

J. Med. Chem. 33 (1990) 311-317.

838

[36]

TE D

835

N. Masuda, O. Yamamoto, M. Fujii, T. Ohgami, J. Fujiyasu, T. Kontani, A. Moritomo, M. Orita, H. Kurihara, H. Koga, H. Nakahara, S. Kageyama, M. Ohta, H.

840

Inoue, T. Hatta, H. Suzuki, K. Sudo, Y. Shimizu, E. Kodama, M. Matsuoka, M.

841

Fujiwara, T. Yokota, S. Shigeta, M. Baba, Studies of nonnucleoside HIV-1 reverse transcriptase inhibitors. Part 1: Design and synthesis of

842

thiazolidenebenzenesulfonamides, Bioorg. Med. Chem. 12 (2004) 6171-6182.

843 844

[37]

H. Karade, B.N. Acharya, M. Sathe, M.P. Kaushik, Design, synthesis, and

antimalarial evaluation of thiazole-derived amino acids, Med. Chem. Res. 17 (2008)

845

19-29.

846 847

AC C

EP

839

[38]

D. González Cabrera, F. Douelle, T.-S. Feng, A.T. Nchinda, Y. Younis, K.L. White,

848

Q. Wu, E. Ryan, J.N. Burrows, D. Waterson, M.J. Witty, S. Wittlin, S.A. Charman, K.

849

Chibale, Novel orally active antimalarial thiazoles, J. Med. Chem. 54 (2011) 7713-

850

7719.

26

ACCEPTED MANUSCRIPT 851

[39]

N.U. Güzeldemirci, Ö. Küçükbasmacı, Synthesis and antimicrobial activity evaluation

852

of new 1,2,4-triazoles and 1,3,4-thiadiazoles bearing imidazo[2,1-b]thiazole moiety,

853

Eur. J. Med. Chem. 45 (2010) 63-68.

854

[40]

P. Makam, R. Kankanala, A. Prakash, T. Kannan, 2-(2-Hydrazinyl)thiazole derivatives: Design, synthesis and in vitro antimycobacterial studies, Eur. J. Med.

856

Chem. 69 (2013) 564-567.

857

[41]

P. Makam, P.K. Thakur, T. Kannan, In vitro and in silico antimalarial activity of 2-(2hydrazinyl)thiazole derivatives, Eur. J. Pharm. Sci. 52 (2014) 138-145.

858 859

RI PT

855

[42]

M.P. Gleeson, A. Hersey, D. Montanari, J. Overington, Probing the links between in vitro potency, ADMET and physicochemical parameters, Nat. Rev. Drug Discov. 10

861

(2011) 197-208.

862

[43]

SC

860

S. Khojasteh, H. Wong, C.C.A. Hop, ADME properties and their dependence on physicochemical properties, in: Drug metabolism and pharmacokinetics quick guide,

864

Springer New York, 2011, pp. 165-181.

865

[44]

M AN U

863

C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J. Feeney, Experimental and

866

computational approaches to estimate solubility and permeability in drug discovery

867

and development settings, Adv. Drug Deliver. Rev. 46 (2001) 3-26. [45]

D.F. Veber, S.R. Johnson, H.-Y. Cheng, B.R. Smith, K.W. Ward, K.D. Kopple,

TE D

868 869

Molecular properties that influence the oral bioavailability of drug candidates, J. Med.

870

Chem. 45 (2002) 2615-2623. [46]

[47]

[48]

Discovery Des. 19 (2000) 179-211.

877

[49]

The influence of lipophilicity in drug discovery and design, Expert Opin. Drug

Discov. 7 (2012) 863-875.

879 880

B. Testa, P. Crivori, M. Reist, P.-A. Carrupt, The influence of lipophilicity on the

pharmacokinetic behavior of drugs: Concepts and examples, Perspect. Drug

876

878

Á. Tarcsay, K. Nyíri, G.M. Keserű, Impact of lipophilic efficiency on compound quality, J. Med. Chem. 55 (2012) 1252-1260.

874 875

EP

making in medicinal chemistry, Nat. Rev. Drug Discov. 6 (2007) 881-890.

872 873

P.D. Leeson, B. Springthorpe, The influence of drug-like concepts on decision-

AC C

871

[50]

N. Siddiqui, W. Ahsan, Triazole incorporated thiazoles as a new class of

881

anticonvulsants: Design, synthesis and in vivo screening, Eur. J. Med. Chem. 45

882

(2010) 1536-1543.

883 884

[51]

M.P. Hay, S. Turcotte, J.U. Flanagan, M. Bonnet, D.A. Chan, P.D. Sutphin, P. Nguyen, A.J. Giaccia, W.A. Denny, 4-Pyridylanilinothiazoles that selectively target 27

ACCEPTED MANUSCRIPT 885

von Hippel−Lindau deficient renal cell carcinoma cells by inducing autophagic cell

886

death, J. Med. Chem. 53 (2009) 787-797.

887

[52]

A. Gallardo-Godoy, J. Gever, K.L. Fife, B.M. Silber, S.B. Prusiner, A.R. Renslo, 2-

888

Aminothiazoles as therapeutic leads for prion diseases, J. Med. Chem. 54 (2011)

889

1010-1021. [53]

K. Jain, J. Bariwal, M. Kathiravan, V. Raskar, G. Wankhede, N. Londhe, An efficient

RI PT

890 891

and rapid synthesis of 2-amino-4-arylthiazoles employing microwave irradiation in

892

water, Green and Sustainable Chem. 1 (2011) 36-40 [54]

arylthiazole derivatives, Asian J. Res. Chem. 2 (2009) 440-444.

894 895

R. Basawaraj, M. Shabbir, Synthesis and biological screening of some new series of

[55]

SC

893

M.G. Bursavich, D.P. Parker, J.A. Willardsen, Z.-H. Gao, T. Davis, K. Ostanin, R. Robinson, A. Peterson, D.M. Cimbora, J.-F. Zhu, B. Richards, 2-Anilino-4-aryl-1,3-

897

thiazole inhibitors of valosin-containing protein (VCP or p97), Bioorg. Med. Chem.

898

Lett. 20 (2010) 1677-1679.

899

[56]

M AN U

896

A.K. Sharma, U.H. Sk, M.A. Gimbor, J.A. Hengst, X. Wang, J. Yun, S. Amin,

900

Synthesis and bioactivity of sphingosine kinase inhibitors and their novel aspirinyl

901

conjugated analogs, Eur. J. Med. Chem. 45 (2010) 4149-4156. [57]

R.G. Gentles, K. Grant-Young, S. Hu, Y. Huang, M.A. Poss, C. Andres, T. Fiedler, R.

TE D

902 903

Knox, N. Lodge, C.D. Weaver, D.G. Harden, Initial SAR studies on apamin-

904

displacing 2-aminothiazole blockers of calcium-activated small conductance

905

potassium channels, Bioorg. Med. Chem. Lett. 18 (2008) 5316-5319. [58]

B.M. Silber, S. Rao, K. Fife, A. Gallardo-Godoy, A. Renslo, D. Dalvie, K. Giles, Y.

EP

906

Freyman, M. Elepano, J. Gever, Z. Li, M. Jacobson, Y. Huang, L. Benet, S. Prusiner,

908

Pharmacokinetics and metabolism of 2-aminothiazoles with antiprion activity in mice, Pharmaceut. Res. 30 (2013) 932-950.

909 910

[59]

C. Pascard, Small-molecule crystal structures as a structural basis for drug design,

Acta Crystallogr., Sect. D: Biol. Crystallogr. 51 (1995) 407-417.

911 912

AC C

907

[60]

K.A. Brameld, B. Kuhn, D.C. Reuter, M. Stahl, Small molecule conformational

913

preferences derived from crystal structure data. A medicinal chemistry focused

914

analysis, J. Chem. Inf. Model. 48 (2008) 1-24.

915

[61]

M. Balganesh, S. Kuruppath, N. Marcel, S. Sharma, A. Nair, U. Sharma, Rv1218c, an

916

ABC transporter of Mycobacterium tuberculosis with implications in drug discovery,

917

Antimicrob. Agents Chemother. 54 (2010) 5167-5172.

28

ACCEPTED MANUSCRIPT 918

[62]

G.M. Morris, R. Huey, W. Lindstrom, M.F. Sanner, R.K. Belew, D.S. Goodsell, A.J.

919

Olson, AutoDock 4 and AutoDockTools 4: Automated docking with selective

920

receptor flexibility, J. Comput. Chem. 30 (2009) 2785-2791.

921

AC C

EP

TE D

M AN U

SC

RI PT

922

29

ACCEPTED MANUSCRIPT 923

Figure and Scheme Legends

924

Figure 1. Rational design of the 2-aminothiazole derivatives. Log P is considered as the

926

decisive factor.

927

Figure 2. Single crystal X-ray diffraction results of 7g. (a) The ORTEP diagram with 40%

928

ellipsoid probability (b) The crystal packing diagram along the c-axis and (c) The single helix

929

packing arrangement along the a-axis.

930

Figure 3. Single crystal X-ray diffraction results of 8l. (a) The ORTEP diagram with 40%

931

ellipsoid probability (b) The crystal packing diagram along the a-axis and (c) The single helix

932

packing arrangement along the b-axis.

933

Figure 4. The summary of structure activity relationship studies.

934

Figure 5. The summary of correlation between Log P and in vitro antimycobacterial activity.

935

Figure 6. Hydrogen bond interactions of compound 7n with Mtb KasA protein.

936

Scheme 1. Synthesis of (i) arylthioureas (ii) pyridyl thioureas (iii) (bromoacetyl)pyridines

937

and (iv) 2-aminothiazole derivatives.

AC C

EP

TE D

M AN U

SC

RI PT

925

30

ACCEPTED MANUSCRIPT Table 1. Physico-chemical properties a and antimycobacterial activity of thiazol-2-amine derivatives against Mtb, H37Rv. S. No. Compound Log P H Freely MIC Mol. H Lipinski Polar Weight rule Acceptors Donors Rotatable Surface (µM) violations Area Bonds 1 5.70 349.228 2 1 1 3 24.919 12.5 7a 7b

6.35

363.255

2

1

1

3

7c

6.64

428.124

2

1

1

4

7d

5.54

304.777

2

1

1

5

7e

5.09

302.786

3

2

0

6

7f

4.77

300.358

3

1

7

7g

4.33

298.367

4

2

8

7h

6.48

361.803

5

1

9

7i

5.92

320.392

2

10

7j

7.62

381.835

11

7k

5.80

332.764

12

7l

3.71

271.32

13

7m

4.34

332.226

14

7n

3.27

15

7o

5.42

16

7p

3.71

17

7q

4.34

18

7r

19

7s

20

8a

21

3

24.919

RI PT

2

25

24.919

100

3

24.919

25

3

45.147

100

0

4

34.153

12.5

0

4

54.381

200

1

5

79.97

>200

1

1

3

24.919

12.5

4

1

1

4

70.74

>200

5

1

1

4

83.63

>200

3

1

0

3

37.811

25

3

1

0

3

37.811

6.25

269.329

4

2

0

3

58.039

>200

332.764

5

1

1

4

83.63

>200

271.32

3

1

0

3

37.811

>200

332.226

3

1

0

3

37.811

>200

M AN U

TE D

AC C

EP

SC

3

3.27

269.329

4

2

0

3

58.039

>200

5.42

332.764

5

1

1

4

83.63

>200

4.01

283.356

4

1

0

4

47.045

>200

8b

4.93

332.226

3

1

0

3

37.811

>200

22

8c

5.58

346.253

3

1

0

3

37.811

>200

23

8d

5.87

411.122

3

1

1

3

37.811

>200

24

8e

2.95

254.318

4

1

0

3

50.703

>200

25

8f

3.46

268.345

4

1

0

3

50.703

100

26

8g

3.46

268.345

4

1

0

3

50.703

>200

27

8h

5.89

336.247

3

1

1

3

37.811

>200

28

8i

6.38

425.149

3

1

1

3

37.811

>200

29

8j

6.09

360.28

3

1

1

3

37.811

>200

30

8k

5.71

360.28

3

1

0

3

37.811

>200

31

8l

4.14

297.383

4

1

0

4

47.045

>200

32

8m

5.51

336.247

3

1

1

3

37.811

>200

33

8n

3.08

268.345

4

1

0

3

50.703

100

34

8o

5.29

317.417

3

1

0

3

37.811

>200

EP

TE D

RI PT

M AN U

SC

Calculated from online server http://www.molinspiration.com/cgi-bin/properties

AC C

a

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT Table 2. Library of N-phenyl thiazol-2-amine and N-2-pyridyl thiazol-2-amine derivatives. Category I: N,4-Diphenylthiazol-2-amine derivatives

8a

8f

RI PT

SC

M AN U

7p

7d

7g 7h 7i Category II: N-Phenyl-4-(pyridinyl)thiazol-2-amine derivatives

7l

7m

7n

8b

7j

7o

8g

8c

8d

8e

8h

8i

8j

8k Category IV: N,4-Di(pyridinyl)thiazol-2-amine derivatives

8l

7e

7q 7r 7s Category III: 4-Phenyl-N-(pyridinyl)thiazol-2-amine derivatives

TE D

7k

7c

EP

7f

7b

AC C

7a

8m

8n

8o

ACCEPTED MANUSCRIPT Table 3. Crystal and measurement details of 7g and 8l molecules. Crystal Parameter

7g

8l

C15H12.5ClN2OS

C13H10N4S

CCDC Number

903724

903727

Formula weight

624.58

254.32

Crystal system

Monoclinic

Monoclinic

Crystal morphology

Colourless block

Pale Yellow needle

Crystal size (mm)

0.45 × 0.37 × 0.25

0.42 × 0.14 × 0.14

Space group

C2/c

P21/C

Temperature/K

293

293

Radiation, λ

Mo Kα , 0.71073 Å

Mo Kα , 0.71073 Å

a (Å)

31.9952 (16)

14.612 (3)

b (Å)

11.3729 (6)

c (Å)

7.8990 (4)

14.715 (3)

α (Å)

-

90°

β (Å)

93.304 (4)°

104.79 (2)°

γ (Å)

-

90°

Volume (Å3)

2869.5 (3)

SC 5.5299 (11)

M AN U

TE D

Z

RI PT

Formula

1149.6 (4)

4

4

1292

528.0

Density (Mg m−3)

1.446

1.469

µ (1/mm)

0.41

0.27

θ (min, max)

3.2, 25.0°

2.9, 25.0°

No. Unique refln

2538

2032

No. of parameters

194

163

Rint, wR(F2)

0.022, 0.109

0.075, 0.128

∆ρmin(eÅ-3), ∆ρmax(eÅ-3)

−0.47, 0.39

−0.34, 0.33

GooF

1.02

0.96

AC C

EP

F (000)

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C EP TE D

SC

M AN U

RI PT

Figure 5

AC C EP TE D

SC

M AN U

RI PT

Figure 6

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Research Highlights Library of 2-aminothiazole derivatives were synthesized and characterized.



All the compounds were evaluated against M. tuberculosis, H37Rv, by in vitro assay.



4-Halophenyl at 2nd position of 2-aminothiazole derivatives enhances activity.



2-Pyridyl at 2nd position of 2-aminothiazole derivatives decreases activity.



The MIC and Ki values of compound 7n are 6.5 µM and 0.44 µM respectively.

AC C

EP

TE D

M AN U

SC

RI PT



SPECTRAL ANALYSIS OF

AC C

EP

TE D

M AN U

SC

CATEGORY-I COMPOUNDS

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

7a: 1H NMR (400 MHz, CDCl3) δ

AC C

EP

TE D

7a: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7b: 13C NMR (101 MHz, CDCl3)

M AN U

SC

RI PT

7b: 1H NMR (400 MHz, CDCl3) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7c: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7c: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7d: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7d: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7e: 13C NMR (101 MHz, CDCl3) δ

M AN U

SC

RI PT

7e: 1H NMR (400 MHz, CDCl3) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7f: 13C NMR (101 MHz, CDCl3) δ

M AN U

SC

RI PT

7f: 1H NMR (400 MHz, CDCl3) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

7g: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

7g: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7h: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7h: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

7i: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

7i: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

7j: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

7j: 13C NMR (101 MHz, DMSO) δ

ACCEPTED MANUSCRIPT

OF

AC C

EP

TE D

M AN U

SC

CATEGORY-II COMPOUNDS

RI PT

SPECTRAL ANALYSIS

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7k: 13C NMR (101 MHz, CDCl3) δ

M AN U

SC

RI PT

7k: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7l: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7l: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7m: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7m: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

7n: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

7n: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7o: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7o: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

TE D

M AN U

SC

RI PT

7p: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

7p: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7q: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7q: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

7r: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

7r: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

TE D

M AN U

SC

RI PT

7s: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

7s: 13C NMR (101 MHz, DMSO-d6) δ

SPECTRAL ANALYSIS

SC

OF

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

CATEGORY-III COMPOUNDS

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8a: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8a: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

8b: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

8b: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8c: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8c: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8d: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8d: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

8e: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

8e: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8f: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8f: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8g: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8g: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8h: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8h: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8i: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8i: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

TE D

M AN U

SC

RI PT

8j: 1H NMR (400 MHz, CDCl3 + DMSO-d6) δ

AC C

EP

8j: 13C NMR (101 MHz, DMSO-d6 + CDCl3) δ

ACCEPTED MANUSCRIPT

TE D

M AN U

SC

RI PT

8k: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

8k: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8l: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8l: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8m: 13C NMR (101 MHz, CDCl3) δ

M AN U

SC

RI PT

8m: 1H NMR (400 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

M AN U

SC

RI PT

8n: 1H NMR (400 MHz, DMSO-d6) δ

AC C

EP

TE D

8n: 13C NMR (101 MHz, DMSO-d6) δ

ACCEPTED MANUSCRIPT

AC C

EP

TE D

8o: 13C NMR (101 MHz, DMSO-d6) δ

M AN U

SC

RI PT

8o: 1H NMR (400 MHz, DMSO-d6) δ

2-Aminothiazole derivatives as antimycobacterial agents: Synthesis, characterization, in vitro and in silico studies.

A series of 2-aminothiazole derivatives with a wide range of substitutions at 2-, 4- and 5-positions were designed and synthesized using Hantzsch thia...
10MB Sizes 4 Downloads 11 Views