Synthesis and anti-tumor evaluation of B-ring substituted steroidal 4 pyrazoline derivatives

10
1 3 Synthesis and anti-tumor evaluation of B-ring substituted steroidal 4 pyrazoline derivatives 5 6 7 Shamsuzzaman a,Q1 , Hena Khanam a , Ashraf Mashrai a , Asif Sherwani b , Mohammad Owais b , 8 Nazish Siddiqui c 9 a Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India 10 b Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India 11 c Department of Illmul Advia Ajmal Khan Tibbiya College, Aligarh Muslim University, Aligarh 202002, India 12 13 14 16 article info 17 Article history: 18 Received 13 February 2013 19 Received in revised form 31 August 2013 20 Accepted 7 September 2013 21 Available online xxxx 22 Keywords: 23 Cholest-5-en-7-one 24 Pyrazolines 25 Anticancer 26 MTT 27 SEM 28 29 abstract 30 The synthesis and anti-tumor activity screening of new steroidal derivatives (418) containing pharma- 31 cologically attractive pyrazoline moieties are performed. During in vitro anticancer evaluation, the newly 32 synthesized compounds displayed moderate to good cytotoxicity on cervical and leukemia cancer cell 33 lines. In addition these compounds were found to be nontoxic to normal cell (PBMCs) (IC 50 > 50 lM). 34 The structure–activity relationship is also discussed. The most effective anticancer compound 9 was 35 found to be active with IC 50 value of 10.6 lM. It demonstrated significant antiproliferative influence on 36 Jurkat cell lines. The morphological changes and growth characteristics of HeLa cells treated with com- 37 pound 4 were analyzed by means of SEM. 38 Ó 2013 Published by Elsevier Inc. 39 40 41 1. Introduction 42 Cancer is becoming the biggest health hazard for the world. 43 Despite recent advances in early diagnosis, prevention and ther- 44 apy, cancer still affects millions of people worldwide and is one 45 of the leading causes of death. In 2008, 7.6 million people died 46 from cancer according to the World Health Organization (WHO) 47 and without immediate action, the global number of deaths from 48 cancer will increase by nearly 80% by 2030 [1]. Although cancer 49 chemotherapy has established a new era of molecularly targeted 50 therapeutics, the efficacy of the existing drugs for the treatment 51 of various cancers is rather limited [2], and there is a need to devel- 52 op new therapeutic agents to overcome the limitations with the 53 current therapy. That’s why there is an intense effort in cancer 54 research to design new, potent, selective and less toxic anticancer 55 agents that are capable of rapid destruction of tumor vasculature 56 leading to tumor necrosis and anti-tumor efficacy [3,4]. In vitro 57 studies, using a variety of human cancer cell lines, have been em- 58 ployed to evaluate the effectiveness of new medicinal compounds 59 against these cancers. 60 Steroids are an important class of natural products which have 61 high ability to penetrate cells and bind to nucleus and membrane 62 receptors. They include great variations in structure and play a 63 very important role in life [5,6]. Structurally diverse cytotoxic 64 and cytostatic steroids are very relevant as lead compounds and 65 molecular probes for anticancer drug discovery and cancer 66 molecular mechanisms elucidation. The chemistry of steroids has 67 motivated extensive investigation through decades and a compre- 68 hensive review on the syntheses of novel bioactive steroids has 69 been recently published [7]. The investigation of modified steroid 70 derivatives condensed with various heterocyclic rings has drawn 71 great attention [8]. A variety of steroids with unusual and interest- 72 ing structures have been synthesized and evaluated for their anti- 73 tumor [9,10], antimicrobial [11] and anti-parasitic activities [12]. 74 Hybrid anti-cancer agents, which combine two active compounds 75 in one, such as steroidal alkylators, contain steroidal moiety as bio- 76 logical vectors for anti-tumor agents in order to diminish toxicity 77 and to enhance specificity, were recently demonstrated [13]. Such 78 types of agents attain duplicate effects on cancer cells. These 79 merged molecules may act on multiple therapeutic targets and of- 80 fer the possibility of circumventing drug resistance. In addition, the 81 hybrids may also minimize unwanted side effects and allow for 82 synergic action [14]. 83 Pyrazolines present an interesting group of compounds, which 84 has been known to possess wide spread pharmacological proper- 85 ties [15]. Recently, different authors worldwide have reported anti- 86 tumor, antiproliferative or anticancer potential of pyrazoline 87 derivatives [16–18]. These derivatives are also well known for their 0039-128X/$ - see front matter Ó 2013 Published by Elsevier Inc. http://dx.doi.org/10.1016/j.steroids.2013.09.006 Corresponding author. Tel.: +91 9411003465. E-mail addresses: [email protected], [email protected] (Shamsuzzaman). Steroids xxx (2013) xxx–xxx Contents lists available at ScienceDirect Steroids journal homepage: www.elsevier.com/locate/steroids STE 7455 No. of Pages 10, Model 5G 26 September 2013 Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumor evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids (2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

Transcript of Synthesis and anti-tumor evaluation of B-ring substituted steroidal 4 pyrazoline derivatives

1

3

4

5

6

7 Q1

8

91011

121314

1 6

1718192021

22232425262728

2 9

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

Contents lists available at ScienceDirect

Steroids

journal homepage: www.elsevier .com/locate /s teroids

Synthesis and anti-tumor evaluation of B-ring substituted steroidalpyrazoline derivatives

0039-128X/$ - see front matter � 2013 Published by Elsevier Inc.http://dx.doi.org/10.1016/j.steroids.2013.09.006

⇑ Corresponding author. Tel.: +91 9411003465.E-mail addresses: [email protected], [email protected]

(Shamsuzzaman).

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumor evaluation of B-ring substituted steroidal pyrazoline derivatives. S(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

Shamsuzzaman a,⇑, Hena Khanam a, Ashraf Mashrai a, Asif Sherwani b, Mohammad Owais b,Nazish Siddiqui c

a Department of Chemistry, Aligarh Muslim University, Aligarh 202002, Indiab Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, Indiac Department of Illmul Advia Ajmal Khan Tibbiya College, Aligarh Muslim University, Aligarh 202002, India

a r t i c l e i n f o

303132333435363738

Article history:Received 13 February 2013Received in revised form 31 August 2013Accepted 7 September 2013Available online xxxx

Keywords:Cholest-5-en-7-onePyrazolinesAnticancerMTTSEM

a b s t r a c t

The synthesis and anti-tumor activity screening of new steroidal derivatives (4–18) containing pharma-cologically attractive pyrazoline moieties are performed. During in vitro anticancer evaluation, the newlysynthesized compounds displayed moderate to good cytotoxicity on cervical and leukemia cancer celllines. In addition these compounds were found to be nontoxic to normal cell (PBMCs) (IC50 > 50 lM).The structure–activity relationship is also discussed. The most effective anticancer compound 9 wasfound to be active with IC50 value of 10.6 lM. It demonstrated significant antiproliferative influence onJurkat cell lines. The morphological changes and growth characteristics of HeLa cells treated with com-pound 4 were analyzed by means of SEM.

� 2013 Published by Elsevier Inc.

39

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

1. Introduction

Cancer is becoming the biggest health hazard for the world.Despite recent advances in early diagnosis, prevention and ther-apy, cancer still affects millions of people worldwide and is oneof the leading causes of death. In 2008, 7.6 million people diedfrom cancer according to the World Health Organization (WHO)and without immediate action, the global number of deaths fromcancer will increase by nearly 80% by 2030 [1]. Although cancerchemotherapy has established a new era of molecularly targetedtherapeutics, the efficacy of the existing drugs for the treatmentof various cancers is rather limited [2], and there is a need to devel-op new therapeutic agents to overcome the limitations with thecurrent therapy. That’s why there is an intense effort in cancerresearch to design new, potent, selective and less toxic anticanceragents that are capable of rapid destruction of tumor vasculatureleading to tumor necrosis and anti-tumor efficacy [3,4]. In vitrostudies, using a variety of human cancer cell lines, have been em-ployed to evaluate the effectiveness of new medicinal compoundsagainst these cancers.

Steroids are an important class of natural products which havehigh ability to penetrate cells and bind to nucleus and membrane

84

85

86

87

receptors. They include great variations in structure and play avery important role in life [5,6]. Structurally diverse cytotoxicand cytostatic steroids are very relevant as lead compounds andmolecular probes for anticancer drug discovery and cancermolecular mechanisms elucidation. The chemistry of steroids hasmotivated extensive investigation through decades and a compre-hensive review on the syntheses of novel bioactive steroids hasbeen recently published [7]. The investigation of modified steroidderivatives condensed with various heterocyclic rings has drawngreat attention [8]. A variety of steroids with unusual and interest-ing structures have been synthesized and evaluated for their anti-tumor [9,10], antimicrobial [11] and anti-parasitic activities [12].Hybrid anti-cancer agents, which combine two active compoundsin one, such as steroidal alkylators, contain steroidal moiety as bio-logical vectors for anti-tumor agents in order to diminish toxicityand to enhance specificity, were recently demonstrated [13]. Suchtypes of agents attain duplicate effects on cancer cells. Thesemerged molecules may act on multiple therapeutic targets and of-fer the possibility of circumventing drug resistance. In addition, thehybrids may also minimize unwanted side effects and allow forsynergic action [14].

Pyrazolines present an interesting group of compounds, whichhas been known to possess wide spread pharmacological proper-ties [15]. Recently, different authors worldwide have reported anti-tumor, antiproliferative or anticancer potential of pyrazolinederivatives [16–18]. These derivatives are also well known for their

teroids

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

Compound 4

Compound 7

Compound 9

Compound 18

Fig. 1. Dose-dependent effects of steroidal pyrazolines (4,7,9,18) on cell viability ofHeLa, Jurkat and PBMC cell lines. Data shown are mean ± standard error of at leastthree independent experiments.

2 Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

pronounced anti-inflammatory, analgesic [19], antidepressant [20]and antimicrobial activities [21]. In addition, antidiabetic activitiesof many compounds containing pyrazoline rings have been re-viewed [22]. The above application of steroids as well as pyrazolinederivatives gave us immense confidence to prepare some new ste-roidal pyrazoline derivatives by combining heterocyclic moiety ofpotent cytotoxic activity with steroid skeleton. In continuation ofour programme towards synthesis of new steroidal derivatives[23], we wish to report herein the facile synthesis of new B-ringsubstituted steroidal pyrazoline derivatives 4–18 starting from ste-roidal a, b-unsaturated ketones 1–3. The evaluation of their anti-tumor activity was carried out in vitro against human cancer celllines (HeLa and Jurkat) and normal cells (PBMCs) using MTT assay.HeLa cells treated with compounds 4, 9 and 18 were observed un-der Fluorescence Microscope (Fig. 2).The surface morphology ofthe treated (with compound 4) and untreated fixed HeLa cells werealso studied (Fig 3).

2. Experimental

2.1. Chemistry

2.1.1. GeneralAll glass apparatus were oven-dried prior to use. Chemicals and

solvents used in this study were of ACS grade and used directlywithout additional steps of purification. Melting points were deter-mined on a Kofler apparatus and are uncorrected. The IR spectrawere recorded on KBr pellets with Interspec 2020 FT-IR Spectrom-eter spectro Lab and values are given in cm�1. 1H and 13C NMRspectra were run in CDCl3/DMSO-d6 on a Bruker Avance II 400NMR Spectrometer (operating at 400 MHz for 1H and at 100 MHzfor 13C NMR) with tetramethylsilane (TMS) as internal standardand values are given in parts per million (ppm) (d). Splitting pat-terns are described as singlet (s), doublet (d), triplet (t) and multi-plet (m). Mass spectra were recorded on a JEOL D-300 massspectrometer. Elemental analyses of all the new compounds wererecorded on Perkin Elmer 2400 CHN Elemental Analyzer. Fluores-cence images were observed under Zeiss Imager M2, Gottingen(Germany) Fluorescence microscope. Scanning electron micro-graph (SEM) was obtained using JSM 6510LV scanning electronmicroscope (JEOL, Tokyo, Japan) at an accelerating voltage of 10and 20 kV. HPLC analysis was performed by LC-100 HPLC instru-ment. Thin layer chromatography (TLC) plates were coated withsilica gel G and exposed to iodine vapors to check the homogeneityas well as the progress of reaction. Sodium sulfate (anhydrous) wasused as a drying agent.

2.1.2. General procedure for the synthesis of steroidal pyrazolinederivatives 4–18

To a solution of cholest-5-en-7-one 1–3 (1.0 mmol) in dichloro-methane/methanol (1:4) (10 ml), hydrazine hydrate (1.5 mmol)was added followed by acetyl chloride (compounds 7–9)/formicacid (compounds 10–12)/benzoic acid (compounds 13–15)/mer-captoacetic acid (compounds 16–18) (3.5 mmol). The reaction mix-ture was refluxed for 3–11 h. The progress as well as completion ofreaction was monitored by TLC. After completion of the reaction,the reaction mixture was cooled to room temperature. The precip-itate thus obtained was filtered, washed with water, air dried andmonitored through TLC for the purity. Thin layer chromatographyrevealed just a single spot which proved the presence of a singleproduct. For further purification, the product was recrystallizedfrom methanol to give product as solid powder. The compounds4–6 were prepared by the same method without addition of anyacid.

The purity of compounds was analyzed by HPLC (Supplemen-tary Fig. SH). Twenty microliter of each sample were injected into

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

the HPLC analysis system by manual injection after they were dis-solved in chloroform. The mobile phase for separation was a mix-ture of methanol and acetonitrile (60:40, vol/vol), and lasted for10 min; the solvent mixture was maintained at a flow rate of

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

Fig. 2. Micrographs showing effect of steroidal pyrazoline derivatives on HeLa Cells. Micrographs captured after 48 h of incubation in bright field under FluorescenceMicroscope. A = Untreated control, B = Treated with comp 18, C = Treated with comp 9, D = treated with comp 4.

Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx 3

STE 7455 No. of Pages 10, Model 5G

26 September 2013

1.000 mL/min. UV detection was performed simultaneously at230 nm wavelength.

2.1.2.1. 3b-Acetoxy-5a-cholestano-[5,7-c d]-pyrazoline (4).Mp: 203–205 �C; Anal. Calc. for C29H48N2O2: C, 76.27, H, 10.59, N, 6.13.Found: C, 76.45, H, 10.46, N, 6.01; IR (KBr) m cm�1: 3417 (N–H),1740 (C@O), 1244 (C–O), 1641 (C@N); 1H NMR (400 MHz, CDCl3):d 2.17 (1H, s, NH, exchangeable with D2O), 4.70 (1H, m, C3-aH, W1/

2 = 18 Hz, axial), 2.02 (3H, s, OCOCH3), 1.00 (3H, s, C10-CH3), 0.71(3H, s, C13-CH3), 0.92 & 0.85 (other methyl protons); 13C NMR(100 MHz, CDCl3): d 171.0, 153.1, 72.0, 53.2, 49.5, 48.1, 42.4,38.5, 38.1, 37.4, 37.3, 36.2, 35.2, 35.1, 34.5, 27.3, 26.7, 26.6, 26.4,22.8, 22.3, 21.8, 21.6, 20.4, 20.1, 19.6, 17.4, 16.7, 11.2; MS (ESI):m/z 456.37 [M+] (Calcd for C29H48N2O2, 456.71).

2.1.2.2. 3b-Chloro-5a-cholestano-[5,7-c d]-pyrazoline (5).Mp: 194–196 �C; Anal. Calc. for C27H45ClN2: C, 74.87, H, 10.47, N, 6.47.

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

Found: C, 74.67, H, 10.65, N, 6.31; IR (KBr) m cm�1: 3423 (N–H),1640 (C@N), 720 (C–Cl); 1H NMR (400 MHz, CDCl3): d 2.0 (1H, s,NH, exchangeable with D2O), 3.51 (1H, m, C3-aH, W1/2 = 16 Hz, ax-ial), 1.10 (3H, s, C10-CH3), 0.81 (3H, s, C13-CH3), 0.92 & 0.86 (othermethyl protons); 13C NMR (100 MHz, CDCl3): d 152.0, 55.2, 54.8,51.7, 50.0, 45.1, 44.3, 43.5, 40.0, 39.3, 38.7, 37.3, 37.1, 36.2, 35.7,33.2, 28.2, 28.0, 27.4, 23.8, 23.3, 22.8, 22.6, 21.0, 19.0, 17.0, 12.2;MS (ESI): m/z 432.33/434.32 [M+] (Calcd for C27H45ClN2, 433.12/435.12).

2.1.2.3.5a-Cholestano-[5,7-c d]-pyrazoline (6).Mp: 208–210 �C; Anal.Calcd for C27H46N2: C, 81.34, H, 11.63, N, 7.03. Found: C, 81.52, H,11.47, N, 7.19; IR (KBr) m cm�1: 3410 (N–H), 1635 (C@N); 1HNMR (400 MHz, DMSO-d6): d 2.11 (1H, s, NH, exchangeable withD2O), 1.12 (3H, s, C10-CH3), 0.71 (3H, s, C13-CH3), 0.91 & 0.85(other methyl protons); 13C NMR (100 MHz, DMSO-d6): d 156.0,55.7, 54.6, 50.3, 49.7, 43.0, 39.7, 39.5, 39.4, 39.3, 38.1, 36.2, 35.1,

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

Fig. 3. SEM micrographs of surface ultrastructural characteristics of untreated (a) and treated (b) HeLa cells with compound 4.

4 Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

33.3, 28.6, 28.0, 27.6, 27.4, 23.7, 22.8, 22.6, 22.2, 21.1, 20.6, 19.0,17.0, 12.2; MS (ESI): m/z 398.37 [M+] (Calcd for C27H46N2, 398.67).

200

201

202

203

204

205

206

207

208

209

210

2.1.2.4. 20-Acetyl-3b-acetoxy-5a-cholestano-[5,7-c d]-pyrazoline (7).Mp: 225–227 �C; Anal. Calc. for C31H50N2O3: C, 74.65, H, 10.10, N,5.62. Found: C, 74.47, H, 10.26, N, 5.47; IR (KBr) m cm�1: 1662 (CH3-

C@O), 1733 (OCOCH3), 1623 (C@N), 1262 (C–O); 1H NMR (CDCl3,400 MHz): d 2.01(3H, s, CH3CO), 2.02 (3H, s, OCOCH3), 4.67 (1H,m, C3-aH, W1/2 = 17 Hz, axial), 1.13 (3H, s, C10-CH3), 0.71 (3H, s,C13-CH3), 0.92 & 0.85 (other methyl protons). 13C NMR (CDCl3,100 MHz): d 171.0, 165.1, 154.0, 73.0, 53.5, 49.5, 48.3, 42.1, 38.6,38.4, 37.4, 37.3, 37.2, 35.3, 35.1, 34.7, 27.5, 26.9, 26.5, 26.4, 23.1,22.8, 22.5, 21.8, 21.5, 20.4, 20.1, 19.8, 17.6, 16.7, 11.2; MS (ESI):m/z 498.38 [M+] (Calc. for C31H50N2O3, 498.75).

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

2.1.2.5. 20-Acetyl-3b-chloro-5a-cholestano-[5,7-c d]-pyrazoline (8).Mp: 215–217 �C; Anal. Calc. for C29H47ClN2O: C, 73.31, H, 9.97, N,5.90. Found: C, 73.48, H, 9.79, N, 6.05; IR (KBr) m cm�1: 1670 (CH3-

C@O), 1630 (C@N); 1H NMR (CDCl3, 400 MHz): d 2.05 (3H, s, CH3-

CO), 3.49 (1H, m, C3-aH, W1/2 = 16 Hz, axial), 1.12 (3H, s, C10-CH3), 0.74 (3H, s, C13-CH3), 0.91 & 0.86 (other methyl protons).13C NMR (CDCl3, 100 MHz): d 165, 154.0, 54.8, 55.2, 51.7, 50.0,45.3, 44.3, 43.5, 40.0, 39.5, 38.9, 37.1, 36.3, 36.2, 35.7, 33.2, 28.6,28.0, 27.8, 23.8, 23.3, 22.8, 22.6, 21.5, 21.0, 19.0, 17.0, 12.3; MS(ESI): m/z 474.34/476.33 [M+] (Calc. for C29H47ClN2O, 475.15/477.15).

2.1.2.6. 20-acetyl-5a-cholestano-[5,7-c d]-pyrazoline (9).Mp: 222–224 �C; Anal. Calc. for C29H48N2O: C, 79.03, H, 10.98, N, 6.36.

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

Table 1Antiproliferative activity (IC50 ± SD values) of steroids (1–18) in human tumor celllines and normal cell lines. Doxorubicin (Dox) and 5-Fluorouracil (5-Fu) are drugs ofreference.

IC50 (lM)

Compounds HeLa Jurkat PBMC

1 49.7 ± 3.5 45.1 ± 0.7 602 42.1 ± 0.5 44.5 ± 2.6 55.73 44.2 ± 3.2 46.3 ± 3.0 58.94 15.3 ± 0.3 24.4 ± 2.8 50.15 31.1 ± 1.2 34.5 ± 1.4 50.46 31.6 ± 3.6 42.8 ± 0.1 54.17 39.7 ± 3.2 12.8 ± 0.3 52.18 25.1 ± 0.6 23.9 ± 1.1 50.59 22.5 ± 2.1 10.6 ± 0.1 54.7

10 31.2 ± 2.2 25.2 ± 4.1 53.211 30.3 ± 2.1 32.6 ± 2.6 51.712 40.4 ± 3.4 31.9 ± 0.9 50.113 26.4 ± 0.5 14.8 ± 0.9 53.614 26.9 ± 2.6 23.5 ± 1.7 54.315 33.5 ± 1.7 35.1 ± 1.7 53.116 30.4 ± 2.6 19.4 ± 0.7 51.317 21.4 ± 1.3 34.7 ± 1.9 55.018 20.3 ± 0.5 14.8 ± 0.3 52.4Dox 4.1 ± 0.1 4.3 ± 0.4 –5-Fu 8.1 ± 0.3 9.0 ± 0.6 –

Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx 5

STE 7455 No. of Pages 10, Model 5G

26 September 2013

Found: C, 79.19, H, 10.83, N, 6.54; IR (KBr) m cm�1: 1680 (C@O),1631 (C@N); 1H NMR (CDCl3, 400 MHz): d 2.02 (3H, s, CH3CO),1.10 (3H, s, C10-CH3), 0.81 (3H, s, C13-CH3), 0.92 & 0.85 (othermethyl protons). 13C NMR (CDCl3, 100 MHz): d 165.0, 153.0, 55.7,54.6, 50.3, 49.7, 43.0, 39.7, 39.5, 39.4, 39.3, 38.1, 36.2, 35.1, 33.3,28.6, 28.0, 27.6, 27.4, 23.8, 23.7, 22.8, 22.6, 22.2, 21.1, 20.6, 19.0,16.0, 12.2; MS (ESI): m/z 440.38 [M+]. (Calc. for C29H48N2O, 440.71).

2.1.2.7. 20-Formyl-3b-acetoxy-5a-cholestano-[5,7-c d]-pyrazoline(10). Mp: 260–262 �C; Anal. Calcd for C30H48N2O3: C, 74.34, H,9.98, N, 5.78. Found: C, 74.54, H, 9.80, N, 5.95; IR (KBr) m cm�1:1691(HC@O), 1738 (OCOCH3), 1636 (C@N), 1239 (C–O); 1H NMR(CDCl3, 400 MHz): d 6.4 (1H, s, CHO), 4.71 (1H, m, C3-aH, W1/

2 = 17 Hz, axial), 2.04 (3H, s, OCOCH3), 1.12 (3H, s, C10-CH3), 0.71(s, 3H, C13-CH3), 0.92 & 0.85 (other methyl protons). 13C NMR(CDCl3, 100 MHz): d 170.0, 159.0, 151.1, 72.0, 53.5, 49.5, 48.3,42.1, 38.6, 38.4, 37.4, 37.3, 37.2, 35.2, 35.1, 34.7, 27.5, 26.9, 26.5,26.4, 22.8, 22.5, 21.8, 21.5, 20.4, 20.1, 19.8, 17.9, 16.7, 11.2; MS(ESI): m/z 484.37 [M+] (Calc. for C30H48N2O3, 484.72).

2.1.2.8. 20-Formyl-3b-chloro-5a-cholestano-[5,7-c d]-pyrazoline (11).Mp: 198–200 �C; Anal. Calc. for C28H45ClN2O: C, 72.93, H, 9.84, N,6.08. Found: C, 72.75, H, 9.99, N, 5.93; IR (KBr) m cm�1: 1689(HC@O), 1630 (C@N), 725 (C–Cl); 1H NMR (CDCl3, 400 MHz): d6.5 (1H, s, CHO), 2.62 (1H, m, C3-aH, W1/2 = 15 Hz, axial), 1.01(3H, s, C10-CH3), 0.71 (s, 3H, C13-CH3), 0.91 & 0.85 (other methylprotons). 13C NMR (CDCl3, 100 MHz): d 161.1, 151.0, 54.8, 55.2,51.7, 49.1, 45.3, 44.3, 43.5, 40.0, 39.5, 38.9, 37.1, 36.3, 36.2, 35.7,33.2, 28.6, 28.0, 27.8, 23.8, 23.3, 22.8, 22.6, 21.0, 19.0, 17.0, 12.3;MS (ESI): m/z 460.32/462.32 [M+] (Calc. for C28H45ClN2O, 461.13/463.13).

2.1.2.9. 20-Formyl-5a-cholestano-[5,7-c d]-pyrazoline (12).Mp: 218–220 �C; Anal. Calc. for C28H46N2O: C, 78.82, H, 10.87, N, 6.57.Found: C, 78.64, H, 11.00, N, 6.41; IR (KBr) m cm�1: 1690 (HC@O),1631 (C@N); 1H NMR (CDCl3, 400 MHz): d 6.4 (1H, s, CHO), 1.12(3H, s, C10-CH3), 0.71 (3H, s, C13-CH3), 0.91 & 0.85 (other methylprotons). 13C NMR (CDCl3, 100 MHz): d 160.0, 156.1, 55.7, 54.6,50.3, 49.7, 43.0, 39.7, 39.5, 39.4, 39.3, 38.1, 36.2, 35.1, 33.3, 28.6,28.0, 27.6, 27.4, 23.7, 22.8, 22.6, 22.2, 21.1, 20.6, 19.0, 17.8, 12.2;MS (ESI): m/z 426.36 [M+] (Calc. for C28H46N2O, 426.68).

2.1.2.10. 20-Benzoyl-3b-acetoxy-5a-cholestano-[5,7-c d]-pyrazoline(13).Mp: 225–227 �C; Anal. Calc. for C36H52N2O3: C, 77.10, H, 9.35,N, 5.00. Found: C, 77.28, H, 9.18, N, 5.18; IR (KBr) m cm�1: 1690 (C6-

H5C@O), 1736 (OCOCH3), 1633 (C@N), 1244 (C–O), 3104 (C–H, aro-matic); 1H NMR (CDCl3, 400 MHz): d 4.69 (1H, m, C3-aH, W1/

2 = 18 Hz, axial), 7.4–7.9 (5H, m, aromatic), 2.05 (3H, s, OCOCH3),1.14 (3H, s, C10-CH3), 0.81 (3H, s, C13-CH3), 0.92 & 0.85 (othermethyl protons). 13C NMR (CDCl3, 100 MHz): d 170.0, 160.0,155.1, 120.0, 118.5, 112.3, 112.0, 110.2, 100.1, 72.0, 53.5, 49.5,48.3, 42.1, 38.6, 38.4, 37.4, 37.3, 37.2, 35.3, 35.1, 34.7, 27.5, 26.9,26.5, 26.4, 23.1, 22.8, 22.5, 21.8, 20.4, 20.1, 19.8, 17.6, 16.7, 11.2;MS (ESI): m/z 560.40 [M+] (Calc. for C36H52N2O3, 560.82).

2.1.2.11. 20-Benzoyl-3b-chloro-5a-cholestano-[5,7-c d]-pyrazoline(14).Mp: 230–232 �C; Anal. Calc. for C34H49ClN2O: C, 76.01, H, 9.19,N, 5.21. Found: C, 76.18, H, 9.03, N, 5.37; IR (KBr) m cm�1: 1686 (C6-

H5C@O), 1635 (C@N), 3120 (C–H, aromatic), 720 (C–Cl); 1H NMR(CDCl3, 400 MHz): d 3.43 (1H, m, C3-aH, W1/2 = 17 Hz, axial), 7.5–7.9 (5H, m, aromatic), 1.13 (3H, s, C10-CH3), 0.71 (3H, s, C13-CH3), 0.91 & 0.84 (other methyl protons). 13C NMR (CDCl3,100 MHz): d 160.0, 152.0, 130.0, 129.1, 125.9, 125.6, 123.4, 120.0,55.2, 54.8, 51.7, 50.0, 45.3, 44.3, 43.5, 40.0, 39.5, 38.9, 37.1, 36.3,36.2, 35.7, 33.2, 28.6, 28.0, 27.8, 23.8, 22.8, 22.6, 21.5, 21.0, 19.0,

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

17.0, 12.3; MS (ESI): m/z 536.35/538.35 [M+] (Calc. for C34H49ClN2-

O, 537.22/539.22).

2.1.2.12. 20-Benzoyl-5a-cholestano-[5,7-c d]-pyrazoline (15).Mp:214–216 �C; Anal. Calc. for C34H50N2O: C, 81.22, H, 10.02, N, 5.57.Found: C, 81.06, H, 10.18, N, 5.42; IR (KBr) m cm�1: 1670 (C6H5-

C@O), 1637 (C@N), 3056 (C–H, aromatic); 1H NMR (CDCl3,400 MHz): 7.5–7.9 (5H, m, aromatic), 1.12 (3H, s, C10-CH3), 0.72(3H, s, C13-CH3), 0.92 & 0.84 (other methyl protons). 13C NMR(CDCl3, 100 MHz): d 166.0, 155.0, 123.0, 119.3, 116.5, 112.5,109.2, 100.0, 55.7, 54.6, 50.3, 49.7, 43.0, 39.7, 39.5, 39.4, 39.3,38.1, 36.2, 35.1, 33.3, 28.6, 28.0, 27.6, 27.4, 23.7, 22.8, 22.6, 22.2,21.1, 20.6, 19.0, 18.0, 12.2; MS (ESI): m/z 502.39 [M+] (Calc. forC34H50N2O, 502.78).

2.1.2.13. 20-Mercaptoacetyl-3b-acetoxy-5a-cholestano-[5,7-c d]-pyr-azoline (16).Mp: 190–192 �C; Anal. Calc. for C31H50N2O3S: C, 70.14,H, 9.49, N, 5.28. Found: C, 70.34, H, 9.31, N, 5.44; IR (KBr) m cm�1:2540 (SH), 1739 (OCOCH3), 1692 (CH2CON), 1640 (C@N), 1243 (C–O); 1H NMR (CDCl3, 400 MHz): d 6.30 (2H, s, CH2), 4.71 (1H, m, C3-aH, W1/2 = 17 Hz, axial), 2.04 (3H, s, OCOCH3), 1.51 (1H, s, SH), 1.14(3H, s, C10-CH3), 0.71 (3H, s, C13-CH3), 0.92 & 0.85 (other methylprotons). 13C NMR (CDCl3, 100 MHz): d 170.4, 162.2, 152.4, 72.5,54.6, 50.5, 49.3, 43.0, 39.7, 39.5, 38.7, 38.5, 38.4, 38.2, 37.3, 36.2,36.1, 35.7, 28.5, 28.0, 27.5, 27.4, 25.3, 23.9, 22.8, 22.5, 21.4, 20.9,18.9, 17.7, 12.2; MS (ESI): m/z 530.35 [M+]. (Calc. for C31H50N2O3S,530.82).

2.1.2.14. 20-Mercaptoacetyl-3b-chloro-5a-cholestano-[5,7-c d]-pyraz-oline (17).Mp: 148–150 �C; Anal. Calc. for C29H47ClN2OS: C, 68.67,H, 9.34, N, 5.52. Found: C, 68.84, H, 9.18, N, 5.69; IR (KBr) mcm�1: 2530 (SH),1686 (CH2CON), 1633 (C@N), 727 (C–Cl); 1HNMR (CDCl3, 400 MHz): d 6.32 (2H, s, CH2), 3.87 (1H, m, C3-aH,W1/2 = 16 Hz, axial), 1.62 (1H, s, SH), 1.12 (3H, s, C10-CH3), 0.71(3H, s, C13-CH3), 0.91 & 0.85 (other methyl protons). 13C NMR(CDCl3, 100 MHz): d 160.2, 153.1, 54.4, 50.7, 49.0, 47.2, 43.5,39.5, 39.4, 38.3, 38.2, 38.1, 38.0, 36.7, 36.5, 35.7, 28.1, 28.0, 27.4,27.2, 25.5, 23.7, 22.8, 22.6, 21.3, 20.2, 19.0, 17.6, 12.1; MS(ESI):m/z 506.31/508.31 [M+]. (Calc. for C29H47ClN2OS, 507.22/509.22).

2.1.2.15. 20-Mercaptoacetyl-5a-cholestano-[5,7-c d]-pyrazoline (18).Mp: 160–162 �C; Anal. Calc. for C29H48N2OS: C, 73.67, H, 10.23,

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

309

310

311

312

313

314

315

316

X

H

H

H

OX

H

H

H

NHN X

H

H

H

NNC

O

R

NH2NH2.H2O

CH2Cl2/CH3OH(1:4),

Reflux

NH2NH2.H2O/RCOOH

CH2Cl2/CH3OH(1:4), Reflux

X

H

H

H

NNC

O

H3C

NH

2NH

2.H

2O/C

H3C

OC

l

CH

2Cl 2

/CH

3OH

(1:4

),

R

eflu

x

(1-3)

(10-18)

(4-6)

(7-9)

Compound X R Compound X R

1 OAc - 10 OAc H

2 Cl - 11 Cl H

3 H - 12 H H

4 OAc - 13 OAc C6H5

5 Cl - 14 Cl C6H5

6 H - 15 H C6H5

7 OAc - 16 OAc SHCH2

8 Cl - 17 Cl SHCH2

9 H - 18 H SHCH2

Scheme 1. Synthesis of steroidal pyrazoline derivatives (4–18).

Table 2Effect of solvent in the synthesis of steroidal N-substituted pyrazolines 10–12.

Products Solvent

CH3OH CH2Cl2 (CH3)2CHOH CH2Cl2/CH3OH

Timea (h) Yieldb (%) Timea (h) Yieldb (%) Timea (h) Yieldb (%) Timea (h) Yieldb (%)

10 12 60 15 62 17 65 8 7511 15 68 14 70 18 70 11 8512 9 65 8 62 12 67 6 82

a Reaction progress monitored by TLC.b All yields refer to recrystallized products.

6 Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

N, 5.93. Found: C, 73.85, H, 10.07, N, 6.07; IR (KBr) m cm�1: 2532(SH), 1690 (CH2CON), 1631 (C@N); 1H NMR (CDCl3, 400 MHz): d6.27 (2H, s, CH2), 1.40 (1H, s, SH), 1.13 (3H, s,C10-CH3), 0.71 (3H,s, C13-CH3), 0.91 & 0.85 (other methyl protons). 13C NMR (CDCl3,

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

100 MHz): d 160.2, 152.0, 54.3, 50.2, 49.0, 47.5, 43.3, 39.6, 39.5,38.4, 38.2, 38.1, 38.0, 36.4, 36.3, 35.3, 28.2, 28.1, 27.5, 27.1, 25.3,23.7, 22.4, 22.3, 21.1, 20.2, 19.0, 17.5, 12.6; MS (ESI): m/z 472.35[M+] (Calc. for C29H48N2OS, 472.78).

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx 7

STE 7455 No. of Pages 10, Model 5G

26 September 2013

2.2. Cytotoxicity assay

The cytotoxic potential of steroidal pyrazolines as well as start-ing compounds, against two cancer cell lines, viz. HeLa (cervical),Jurkat (leukemia) (obtained from NCCS Pune, Maharashtra) andnormal cells was assessed by determining the number of viablecells surviving after their incubation with drug for stipulated timeperiod using MTT (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy-methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) method [24].The tumor cell lines (HeLa and Jurkat) and normal cells (PBMC)were maintained in RPMI 1640 culture medium supplementedwith 10% heat-inactivated fetal calf serum. The cells were platedat a density of 5 � 104 cells per well in a 96-well plate, and cul-tured for 24 h at 37 �C. Stock solutions of the synthesized steroidswere prepared in 1:1 mixture of DMSO and THF [25]. The cellswere subsequently exposed to drugs. The plates were incubatedfor 48 h, and cell proliferation was measured by adding 20 lL ofMTT dye (5 mg/mL in phosphate-buffered saline) per well. Theplates were incubated for a further 4 h at 37 �C in a humidifiedchamber containing 5% CO2. Formazan crystals formed due toreduction of dye by viable cells in each well were dissolved in150 lL dimethyl sulfoxide, and absorbance was read at 570 nm.The absorption values were expressed as the cell viability (%),according to the control group as 100%. Assays were performedin triplicate on three independent experiments. The concentrationrequired for 50% inhibition of cell viability (IC50) was calculatedusing the software ‘‘Prism 3.0’’.

2.2.1. Blood peripheral mononuclear cell isolationFresh blood (20–15 mL) was kindly provided by Blood bank

Jawahar Lal Nehru Medical College, AMU Aligarh. The blood samplewas diluted with the same volume of PBS. After that, the dilutedblood sample was carefully layered on Ficoll-Histopaque. The mix-ture was centrifuged under at 400g for 30 min at 20–22 �C. Theundisturbed lymphocyte layer was carefully transferred out. Thelymphocyte was washed and pelleted down with three volumesof PBS for twice and resuspended RPMI-1640 media with antibioticand antimycotic solution 10%, v/v fetal calf serum (FCS). Cell count-ing was performed to determine the PBMC cell number with equalvolume of trypan blue [26].

2.2.2. Fluorescence Microscopy/Scanning Electron MicroscopyThe HeLa cell line was maintained in RPMI 1640 culture med-

ium supplemented with 10% heat-inactivated fetal calf serum.The cells were plated at a density of 104 cells on glass cover slips,and cultured for 24 h at 37 �C. These were subsequently exposedwith compounds for 48 h. Cells were fixed by 2% paraformaldehydefor 2 h followed by washing with HBSS. The HeLa cells treated withcompounds 4, 9 and 18 were observed under Fluorescence Micro-scope (Fig. 2). The surface morphology of the treated (with com-pound 4) and untreated fixed HeLa cells are shown in Fig. 3.

426

427

428

429

430

431

432

433

434

435

436

437

438

439

3. Results and discussion

3.1. Chemistry

The generation of molecules/molecular assemblies possessingwell defined biological functions remains an extremely challengingtask. In the view of significance of different nitrogen containingsteroidal heterocycles, a series of some new N-substituted steroi-dal pyrazoline derivatives have been rationally designed and syn-thesized and evaluated as potential anticancer agents. Thestarting materials 3b-acetoxycholest-5-en-7-one 1, 3b-chlorocho-lest-5-en-7-one 2 and cholest-5-en-7-one 3 were synthesized byknown literature method [27]. These were allowed to react with

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

hydrazine hydrate and acetyl chloride/formic acid/benzoic acid/mercaptoacetic acid in dichloromethane/methanol (1:4) under re-flux conditions for the synthesis of new types of steroids attachedto nitrogen heterocycles 4–18 (Scheme 1). To determine theoptimal reaction conditions, nature of solvents were investigated(Table 2). Initially, the reaction was carried out in methanol (onlycompounds 10–12 were tested as model reaction) but, unfortu-nately, the expected products were not observed in good yield.Similar results were found with CH2Cl2. The use of isopropanol in-creased the chemical yield but, reduced the reaction rate. When weused dichloromethane/methanol (1:4) mixture as solvent, the rateas well as yield of products was improved. We applied this solventsystem for other compounds also. Interestingly, better chemicalyields and fast reaction rate were examined. Spectral data for allthe compounds (4–18) can be found in Supplementary information(Figs. S1–S30).

The structures of the target compounds, 4–18 were readilyidentified by their correct elemental analyses and compatible IR,1H NMR, 13C NMR and MS spectral data. All the analytical and spec-tral data of compounds 4–18 were in accordance with the pro-posed structure. The diagnostic signals at 3410–3423 cm�1 and1635–1641 appeared in the IR spectra of compounds 4–6 wereattributed to N–H and C@N stretching vibrations respectively.The 1H NMR spectra of compounds displayed singlet, at d = 2.0–2.17 ppm, due to N–H proton. The IR spectra of compounds 10–12 showed characteristic absorption peaks at 1689–1691 (HC@O)and 1630–1636 cm�1 (C@N). The 1H NMR spectra of compoundsrevealed the presence of singlet at d 6.4–6.5 which is characteristicfor the formyl proton. The signal at d 159–161 ppm in 13C NMRspectra further supported the presence of formyl group while peakat 150.9–156.7 was due to C@N function. Other compounds werecharacterized in the same way.

A conceivable mechanism for the synthesis of steroidal pyrazo-lines 4–18 is represented in Scheme 2 [28]. a, b-unsaturated ke-tones react with hydrazine hydrate to furnish the intermediate‘c’, which has been trapped by t-butyl carbazate (SupplementaryFig. S). This intermediate reacts with acid/acetyl chloride to endowthe corresponding products 4–18.

We confirmed the general pattern of reaction by reacting 3-methyl-2-cyclohexenone with NH2NH2�H2O,NH2NH2�H2O/CH3-

COCl,NH2NH2�H2O/HCOOH,NH2NH2�H2O/C6H5COOH and NH2NH2-

�H2O/SHCH2COOH (Supplementary Fig. Si–Svi).The obtainedproducts (compounds a–e) were also screened against cancer celllines (HeLa, Jurkat) and normal cell lines (PBMC) (SupplementaryTable S).

3.1.1. StereochemistryThe stereochemical assignation of C5–N bond has been estab-

lished on the basis of mechanism as well as on NMR spectral anal-ysis of the compounds. During the course of reaction, thenucleophilic attack of N of the reagent at C-5 does occur preferablyfrom less hindered (a) side because of the steric encumbrance im-posed by axial (b) methyl group at C-10, resulting axial (a) orienta-tion of C5–N bond and trans to C10-axial methyl group (A/B ringjunction trans) [23a]. In addition the ring fusion stereochemistryin angularly methylated six-membered ring compounds can bedetermined by NMR spectroscopy. The half band width (W1/2) val-ues of C3-axial proton in the 1H NMR spectra of the synthesizedcompounds clearly suggest that A/B ring junction is trans [29].13C NMR values of C-19 are strongly dependent on the ring fusionstereochemistry. The cis and trans steroids differ most significantlyat C-19 and this signal will surely characterize the nature of thering junction [30]. In the compounds 4–18, C-19 chemical shiftvalues were observed in the range of 16–18 ppm, which is consis-tent with the values obtained for trans steroids [31].

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

X

H

H

H

O

X

H

H

H

N

X

H

H

H

OH

NH2-NH2

+

HNNH2

X

H

H

H

O

HN NH

H

X

H

H

H

OH

HN N H

-H2O

H2N

X

H

H

H

HN N

Path a Path b

-H2O

Intermediate c

X

H

H

H

N NC

O

RCH3/

RCOOH/ CH3COCl

Scheme 2. A provisional mechanism for the synthesis of steroidal pyrazoline derivatives (4–18).

8 Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

3.2. Anticancer activity and structure–activity relationship

To gain insight on how modifications on ring-B can affect cyto-toxicity, 15 steroidal pyrazoline derivatives with a variety of func-tionalities attached to N, namely, hydrogen, formyl, acetyl, benzoyland mercaptoacetyl, were synthesized and assayed in vitro forcytotoxicity in human cancer and normal cell lines, using theMTT assay. The cancer cells encompassed HeLa (cervical cancercells) and Jurkat (leukemia) while PBMCs were used as normalcells. A period of 48 h of drug exposure was chosen to test cytotox-icity. Doxorubicin (Dox) and 5-Fluorouracil (5-Fu) were used ascytotoxic drugs of reference. The cytotoxicity (IC50) of the synthe-sized steroidal pyrazoline derivatives along with starting steroid,against cancer lines (HeLa and Jurkat) as well as normal cells aredetailed in Table 1, whereas the curves of dose-dependent effects

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

of most active compounds (4,7,9 and 18) are displayed in Fig. 1and remaining are given in Supplementary data. A number of cor-relations can be made from the data given in the Table 1. It is evi-dent from the IC50 values, that all the compounds showedmoderate to good activity while compounds 4, 7, 9, 13, 16 and18 elicited a marked inhibitory activity (IC50 < 19 lM) against boththe cell lines. All the compounds were found to be nontoxic to nor-mal cells (IC50 > 50 lM). Its noteworthy point that compound 4was found to be specific against HeLa while compounds 7, 9, 13,16 and 18 showed selectivity towards Jurkat cells. Substituent at3b-position as well as groups attached to N of the pyrazoline ringplayed a crucial role in determining activity. It is manifested fromthe data that acetoxy group at 3b-position impart greater activity(compound 4; HeLa, 10, 7 and 13; Jurkat) as compared to H andCl (compound 9 is unexpectedly more potent than 7). But presence

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

469

470

471

472

473

474

475

476

477

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511512513514515516517518519520521522523524525526527

528529530531532533534535536537538539540541542543544545546547548549550551

Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx 9

STE 7455 No. of Pages 10, Model 5G

26 September 2013

of bulky substituent attached to pyrazoline ring nullified this effectand D5 derivatives were found to be more active (compounds 9and 18). Nature of the substituent over the pyrazoline ring alsoinfluenced the relative toxicity. This could be attributed to theirdifferences in either polarity which changes their lipophilicity orthe conformation which alters the target protein binding proper-ties present within the cell or on the cell membrane. HeLa cellstreated with compounds 4, 9 and 18 were observed under Fluores-cence Microscope (Fig. 2). These images clearly designated reduc-tion in cancer cell count, thus showing cytotoxicity of abovementioned steroids. Fig. 3 shows SEM micrograph of untreatedHeLa cells (a) and HeLa cells treated (b) with compound 4. The con-torted-looking state of the treated cells showed distinct morpho-logical changes corresponding to typical apoptosis, includingcellular blebbing and the formation of apoptotic bodies. [32]. Itcould be concluded that newly synthesized steroidal pyrazolineswere able to produce distinctive morphological features of celldeath that might correspond to apoptosis.

552553554555556557558559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610

4. Conclusion

In summary, we have demonstrated an efficient, expedient andconvenient approach for the synthesis of steroidal pyrazolinederivatives. Moreover, fast reaction rate, simple experimentationand better yields of the products are the advantages of this reac-tion. The investigated compounds 4, 7, 9, 13, 16 and 18 exertedinteresting antiproliferative behavior by showing low values ofinhibition count (IC50). In particular, compounds 9 (IC50 = 10.6 lM)and 7 (IC50 = 12.8 lM) showed a better cytotoxic profile among allthe tested compounds and were found to be potent inducer of celldeath in cultured Jurkat cell lines. Furthermore, all the compoundswere found to be non toxic to normal cell lines (PBMC).

Acknowledgements

We sincerely thank, the chairman, Department of Chemistry,Aligarh Muslim University, Aligarh for providing necessary re-search facilities. HK acknowledges UGC, New Delhi India for pro-viding BSR fellowship (R. No. Acad/D-742/MR). Authors alsothank the SAIF, Punjab University Chandigarh for providing spec-tral data.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, inthe online version, at http://dx.doi.org/10.1016/j.steroids.2013.09.006.

References

[1] Burden: Mortality, Morbidity and Risk Factors. In Global Status Report onNoncommunicable Diseases 2010. Description of the Global Burden of NCDs,Their Risk Factors and Determinants; World Health Organization: Geneva,Switzerland, 2011; pp. 9–31.

[2] He J, Wang X, Zhao X, Liang Y, He H, Fu L. Synthesis and antitumor activity ofnovel quinazoline derivatives containing thiosemicarbazide moiety. Eur J MedChem 2012;54:925–30.

[3] Zhou BBS, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. NatRev Drug Dis 2009;8:806–23.

[4] Havrylyuk D, Zimenkovsky B, Vasylenko O, Gzella A, Lesyk R. Synthesis of new4-thiazolidinone-, pyrazoline-, and isatin-based conjugates with promisingantitumor activity. J Med Chem 2012;55:8630–41.

[5] Festi D, Montagnani M, Azzaroli F, Lodato F, Mazzella G, Roda A, Di Biase AR,Roda E, Simoni P, Colecchia A. Clinical efficacy and effectiveness ofursodeoxycholic acid in cholestatic liver diseases. Curr Clin Pharmacol2007;2:155–77.

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

[6] Ifere GO, Barr E, Equan A, Gordon K, Singh UP, Chaudhary J, Igietseme JU,Ananaba GA. Differential effects of cholesterol and phytosterols on cellproliferation, apoptosis and expression of a prostate specific gene in prostatecancer cell lines. Cancer Detect Prev 2009;32:319–28.

[7] Hanson JR. Steroids: partial synthesis in medicinal chemistry. Nat Prod Rep2010;27:887–99.

[8] Mohamed NR, Elmegeed GA, Abdelhalim MM, Rady HM. Facile synthesis andin vitro cytotoxic evaluation of novel thiadiazole, pyrazole, and dithiole-androstane derivatives. J Phosphorus Sulfur Silicon 2010;185:848–56.

[9] Salvador JAR, Carvalho JFS, Neves MAC, Silvestre SM, Leitão AJ, Silva MMC,Melo MLS. Anticancer steroids: linking natural and semi-synthetic compounds.Nat Prod Rep 2013;30:324–74.

[10] Djurendic EA, Sakac MN, Zavis MP, Gakovic AR, Canadi JJ, Andric SA, KlisuricOR, Kojic VV, Bogdanovic GM, Gasi KMP. Synthesis and biological evaluation ofsome new A, B-ring modified steroidal d-lactones. Steroids 2008;73:681–8.

[11] (a) Burbiel J, Bracher F. Azasteroids as antifungals. Steroids 2003;68:587–94;(b) Gogoi S, Shekarrao K, Duarah A, Bora TC, Gogoi S, Boruah RC. A microwavepromoted solvent-free approach to steroidal quinolines and their in vitroevaluation for antimicrobial activities. Steroids 2012;77:1438–45.

[12] Gros L, Lorente SO, Jimenez CJ, Yardley V, Rattray L, Wharton H, Little S, CroftSL, Ruiz-Perez LM, Gonzalez-Pacanowska D, Gilbert IH. Evaluation ofazasterols as anti-parasitics. J Med Chem 2006;49:6094–103.

[13] Trafalis DTP, Geromichalos GD, Koukoulitsa C, Papageorgiou A, Karamanakos P,Camoutsis C. Lactandrate: a D-homo-aza-androsterone alkylator in thetreatment of breast cancer. Breast Cancer Res Treat 2006;97:17–31.

[14] El-Far M, Elmegeed GA, Eskander EF, Rady HM, Tantawy MA. Novel modifiedsteroid derivatives of androstanolone as chemotherapeutic anticancer agents.Eur J Med Chem 2009;44:3936–46.

[15] Zitouni GT, Chevallet P, Kilic FS, Erol K. Synthesis of some thiazolyl-pyrazolinederivatives and preliminary investigation of their hypotensive activity. Eur JMed Chem 2000;35:635–41.

[16] Johnson M, Younglove B, Lee L, LeBlanc R, Holt Jr H, Hills P, Mackay H, Brown T,Mooberry SL, Lee M. Design, synthesis, and biological testing of pyrazolinederivatives of combretastatin-A4. Bioorg Med Chem Lett 2007;17:5897–901.

[17] Rostom SAF. Synthesis and in vitro antitumor evaluation of some indeno [1,2-c]pyrazol(in)es substituted with sulfonamide, sulfonylurea(-thiourea)pharmacophores, and some derived thiazole ring systems. Bioorg Med Chem2006;14:6475–85.

[18] Banday AH, Mir BP, Lone IH, Suri KA, Kumar HMS. Studies on novel D-ringsubstituted steroidal pyrazolines as potential anticancer agents. Steroids2010;75:805–9.

[19] Amir M, Kumar H, Khan SA. Synthesis and pharmacological evaluation ofpyrazoline derivatives as new anti-inflammatory and analgesic agents. BioorgMed Chem Lett 2008;18:918–22.

[20] Johnson M, Younglove B, Lee L, LeBlanc R, Holt Jr H, Hills P, Mackay H, Brown T,Mooberry SL, Lee M. Design, synthesis, and biological testing of pyrazolinederivatives of combretastatin-A4. Bioorg Med Chem Lett 2007;17:5897–901.

[21] Sahu SK, Banerjee M, Samantray A, Behera C, Azam MA. Synthesis, analgesic,anti-inflammatory and antimicrobial activities of some novel pyrazolinederivatives. Trop J Pharm Res 2008;7:961–8.

[22] Ahn JH, Kim HM, Jung SH, Kang SK, Kim KR, Rhee SD, Yang SD, Cheon HG, KimSS. Synthesis and DP-IV inhibition of cyano-pyrazoline derivatives as potentanti-diabetic agents. Bioorg Med Chem Lett 2004;14:4461–5.

[23] (a) Shamsuzzaman, Khanam H, Mashrai A, Siddiqui N. Construction of novelsteroidal isoxazolidinone derivatives under Vilsmeier–Haack conditions.Tetrahedron Lett 2013;54:874–7;Shamsuzzaman, Khan MS, Alam M, Tabassum Z, Ahmad A, Khan AU. Synthesis,antibacterial and antifungal activities of 6,5 fused steroidal oxazoles incholestane series. Eur. J. Med. Chem 2010;45:1094–7;Shamsuzzaman, Ahmad S, Khan BZ, Shafiullah. A convenient method for thesynthesis of 3b-hydroxy-4-en-6-one steroids. J. Org. Chem 1991;56(5):1936–7.

[24] Mosmann T. Rapid colorimetric assay for cellular growth and survival:application to proliferation and cytotoxicity assays. J Immunol Methods1983;65(1–2):55–63. http://dx.doi.org/10.1016/0022-1759(83)90303-4.

[25] Carvalho JFS, Silva MMC, Moreira JN, Simoes S, Melo MLS. Selectivecytotoxicity of oxysterols through structural modulation on rings A and B.Synthesis, in vitro evaluation, and SAR. J Med Chem 2011;54:6375–93.

[26] Yeap SK, Alitheen NB, Ali AM, Omar AR, Raha AR, Suraini AA, Muhajir AH. Effectof Rhaphidophora korthalsii methanol extract on human peripheral bloodmononuclear cell (PBMC) proliferation and cytolytic activity toward HepG2. JEthnopharmacol 2007;114:406–11.

[27] (a) Dauben WG, Takemura KH. A Study of the mechanism of conversion ofacetate to cholesterol via squalene. J Am Chem Soc 1953;75:6302–4;(b) Fieser LF, Fieser M, Chakravarti RN. a-Spinasterol. J Am Chem Soc1949;71:2226–30;Milburn AH, Truter EV. The components of wool wax. Part III⁄ 7-oxocho1esterol and the alleged presence of cholestanol. J. Chem. Soc1956:1736–9.

[28] Huang YR, Katzenellenbogen JA. Regioselective synthesis of 1,3,5-triaryl-4-alkylpyrazoles: novel ligands for the estrogen receptor. Org. Lett 2000;2(18):2833–6.

[29] Hassner A, Heathcock C. Reaction of nitrosyl chloride with steroid 5-Enes.Nuclear Magnetic Resonance as a stereochemical tool in steroids. J Org Chem1964;29:1350–5.

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids

611612613614615

616617618619

620

10 Shamsuzzaman et al. / Steroids xxx (2013) xxx–xxx

STE 7455 No. of Pages 10, Model 5G

26 September 2013

[30] Leibfritz D, Roberts JD. Nuclear magnetic resonance spectroscopy. Carbon-13spectra of cholic acids and hydrocarbons included in sodium desoxycholatesolutions. J. Am. Chem. Soc 1973:4996–5003.

[31] Iida T, Ogawa S, Hosoi K, Makino M, Fujimoto Y, Goto T, Mano N, Goto J,Hofmann AF. Regioselective oxyfunctionalization of unactivated carbons in

Please cite this article in press as: Shamsuzzaman et al. Synthesis and anti-tumo(2013), http://dx.doi.org/10.1016/j.steroids.2013.09.006

steroids by a model of cytochrome P-450: Osmiumporphyrin complex/tert-butyl hydroperoxide system. J Org Chem 2007;72:823–30.

[32] Capua CJ, Hopson NP, Stewart CM, Johnston GR, O’Neill KL, Schaalje GB, LeeCM, Booth GM. Cytotoxicity of Atriplex confertifolia. J Toxicol 2010.

r evaluation of B-ring substituted steroidal pyrazoline derivatives. Steroids