Accepted Manuscript Title: Structure elucidation of a process-related impurity of dapoxetine Author: Andr´as Darcsi Gerg˝o T´oth J´ozsef K¨ok¨osi Szabolcs B´eni PII: DOI: Reference:

S0731-7085(14)00184-8 http://dx.doi.org/doi:10.1016/j.jpba.2014.04.002 PBA 9528

To appear in:

Journal of Pharmaceutical and Biomedical Analysis

Received date: Revised date: Accepted date:

18-2-2014 31-3-2014 3-4-2014

Please cite this article as: http://dx.doi.org/10.1016/j.jpba.2014.04.002 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.

1

Structure elucidation of a process-related impurity of dapoxetine

2 András Darcsi, Gergő Tóth, József Kökösi, Szabolcs Béni*

3 4

Department of Pharmaceutical Chemistry, Semmelweis University

6

H-1092 Hőgyes Endre street 9. Budapest, Hungary

7

cr

8 9

us

10 11

an

12 13 14

M

15 16

ip t

5

Highlights

d

17! ! A novel dapoxetine by-product has been identified using NMR and MS. 18! ! The mechanism for the impurity formation was proposed and also confirmed by synthesis. 20 21 22 23 24

Ac ce p

te

19! ! The separation of dapoxetine and the process-related impurities was accomplished by HPLC.

25

*

26

Dr. Szabolcs Béni

27

Tel.: +36 1 217 0891; fax: +36 1 217 0891; E-mail address:

28

[email protected]

Corresponding author:

29

1 Page 1 of 18

Abstract

30

Unknown by-product associated with the synthesis of dapoxetine was isolated. The structure

31

elucidation of this new compound using accurate mass data and NMR spectroscopy is

32

presented herein. The unambiguous resonance assignment concluded to the formation of a

33

tricyclic compound 4-phenyl-2H,3H,4H-naphtho[1,2-b]pyran, a new impurity of dapoxetine

34

which has never been reported previously. A proposed mechanism for the formation of the

35

new carbon-carbon bond is discussed. For the separation of dapoxetine and the process-

36

related impurities, a gradient HPLC method was developed.

cr

37

ip t

29

Keywords: Priligy, impurity profiling, NMR, HPLC-DAD, electrophilic aromatic

39

substitution, ring closure mechanism

us

38

41

an

40 Highlights

42! ! A novel dapoxetine by-product has been identified using NMR and MS.

M

43! ! The mechanism for the impurity formation was proposed and also confirmed by synthesis. 44! ! The separation of dapoxetine and the process-related impurities was accomplished by HPLC.

Ac ce p

te

d

45

2 Page 2 of 18

46 47 1.

1. Introduction

48

Dapoxetine

49

hydrochloride, Priligy®) is a novel short acting selective serotonin reuptake inhibitor (SSRI)

50

that is being developed specifically as an on-demand oral treatment of premature ejaculation

51

with a unique pharmacokinetic profile [1]. Dpx attains its peak plasma concentration in about

52

1.5 hours after dosing which is much faster than conventional SSRIs and by 24 hours the

53

plasma concentration decreases to approximately 5% of the peak concentration. These

54

pharmacokinetic properties make Dpx an excellent candidate for on-demand treatment of

55

premature ejaculation. The eutomer (S)-Dpx is 3.5 times more potent SSRI than (R)-Dpx, that

56

is why Dpx is marketed as a single enantiomer drug [2]. A wide range of synthetic procedures

57

were developed to synthesize racemic and enantiopure Dpx [3-7].

58

Our previous study reported a robust, sensitive and validated method for the chiral separation

59

of Dpx enantiomers via cyclodextrin-modified capillary electrophoresis [7] along with a

60

synthetic procedure for racemic Dpx following the main literature methods. In this

61

communication, the identification of a by-product is discussed, which was formed in the last

62

step of a common synthetic pathway using in situ mesylation for dapoxetine synthesis [6, 8-

63

15] (Figure 1.).

64

This well-characterized and widely accepted scheme for Dpx synthesis utilizes mesylate as an

65

excellent leaving group in nucleophilic substitution reactions. The mesylate intermediate (5)

66

is formed in situ and usually converted to Dpx without isolation. There is a single reference

67

reporting the characteristics of this intermediate [16], however the isolation of this compound

68

is missing. Several attempts failed to isolate compound 5, but provided an unknown by-

69

product.

70

The goal of this communication is the structure elucidation of this new by-product using

71

NMR spectroscopy and mass spectrometry. Following the structural characterization of this

72

impurity, a mechanism for the formation was also proposed. For the separation of Dpx and the

73

process-related impurities, a gradient HPLC method was developed.

(S)-N,N-dimethyl[3-(naphthalen-1-yloxy)-1-phenylpropyl]amine

Ac ce p

te

d

M

an

us

cr

ip t

(Dpx),

74 75

2.

Materials and methods

76

2.1.

Instrumentation

77

All NMR experiments were carried out on a 600 MHz Varian DDR NMR spectrometer

78

equipped with a 5 mm inverse-detection gradient (IDPFG) probehead. Standard pulse

79

sequences and processing routines available in VnmrJ 3.2 C/Chempack 5.1 were used for 3 Page 3 of 18

80

structure identifications. The complete resonance assignments were established from direct

81

1

82

gCOSY, 1H–13C gHSQCAD (J = 140 Hz), 1H–13C gHMBCAD (J = 8 Hz) experiments,

83

respectively. The probe temperature was maintained at 298 K and standard 5 mm NMR tubes

84

were used. The 1H chemical shifts were referenced to TMS (0.00 ppm) while

85

shifts were referenced to the applied NMR solvent CDCl3 (77.16 ppm).

86

The accurate mass of the products were determined with an Agilent 6230 time-of-flight mass

87

spectrometer. Samples were introduced by the Agilent 1260 Infinity LC system, the mass

88

spectrometer was operated in conjunction with a JetStream (ESI) ion source in positive ion

89

mode. Reference masses of m/z 121.050873 and 922.009798 were used to calibrate the mass

90

axis during analysis. Mass spectra were processed using Agilent MassHunter B.02.00

91

software.

92

HPLC-DAD analyses were performed using an Agilent 1260 Infinity LC apparatus. An

93

Agilent Zorbax Eclipse Plus C18, 100 mm x 4.6 mm i.d. (3.5 μm particle size) column was

94

applied. The mobile phase consisted of methanol and water with 0.1 v/v% formic acid using

95

the following gradient program: 0 min. 50% MeOH, 4 min. 50% MeOH, 6 min. 90% MeOH,

96

10.0 min. 90% MeOH, 11 min. 50% MeOH, 15 min 50% MeOH. The flow rate was set to 1

97

ml/min, while the column temperature was kept at 50°C. The UV spectra were recorded

98

ranging from 200-400 nm.

H–13C, long-range 1H–13C, and scalar spin–spin connectivities using 1D 1H,

13

C chemical

2.2.

101

All reagents for synthesis, HPLC grade solvents used for LC-MS analyses and CDCl3 (99.8

102

atom% D) for NMR were purchased from Sigma-Aldrich. Water was produced by a Millipore

103

Milli-Q Direct 8 water purifying system. TLC was performed using precoated Silica gel 60

104

F254 TLC plates and visualized with ultraviolet light at 254 nm. For column chromatography,

105

the 40-63 ! m silica was used.

Ac ce p

100

106

Chemicals

te

99

d

M

an

us

cr

ip t

13

C, 1H–1H

107

2.3.

Synthesis of 3-chloro-1-phenyl-1-propanol (3)

108

To the solution of 3-chloropropriophenone (1 g; 6 mmol) in THF (14 ml) and water (1 ml),

109

NaBH4 (0.27 g; 7 mmol) was added at 0°C. The mixture was stirred for 24 h at room

110

temperature.

111

After completion (checked by TLC using CH2Cl2, starting material Rf = 0.77, product Rf =

112

0.48), dilute acetic acid (12%) was added slowly to the reaction mixture with stirring at the

113

same temperature to set the pH to 4.5. The crude mixture was treated with water (10 ml), 4 Page 4 of 18

114

extracted with ethyl acetate (2 x 150 ml). The separated organic layers were combined and

115

washed with 5 % NaHCO3 solution. The organic layer was washed with water and dried over

116

sodium sulfate and filtered, followed by solvent evaporation.

117

The crude product was the racemic 3-chloro-1-phenyl-1-propanol (3, 1.02 g, 99%) as yellow

118

oil and subsequently crystallized overnight.

119

1

120

8.5, 4.7 Hz, 1H), 3.78 – 3.70 (m, 1H), 3.59 – 3.53 (m, 1H), 2.28 – 2.20 (m, 1H), 2.13 – 2.06

121

(m, 1H). 13C NMR (CDCl3, 151 MHz): δ(ppm): 143.83, 128.80, 128.05, 125.90, 71.47, 41.85,

122

41.58. HRMS: calc. [M+Na]+ 193.0391, found [M+Na]+ 193.0396.

us

123

cr

ip t

H NMR (CDCl3, 600 MHz): δ(ppm): 7.39 – 7.34 (m, 4H), 7.33 – 7.29 (m, 1H), 4.94 (dd, J =

2.4.

Synthesis of 3-(1-naphthalenyloxy)-1-pheny-l-propanol (4)

125

Operating under inert atmosphere of nitrogen gas, to a mixture of DMF (2.3 ml) and 60%

126

sodium hydride in mineral oil (0.12 g, 2.9 mmol) at 0° C in an ice bath a solution of 1-

127

naphthol (0.43 g, 2.9 mmol) of DMF (2.3 ml) was added dropwise. The reaction mixture was

128

stirred at 0° C for two hours and a solution of 3 (0.53 g, 3.1 mmol) in DMF (2.1 ml) was

129

added. After stirring overnight at room temperature and completion (checked by TLC using n-

130

hexane and EtOAc (9:1, v/v), starting material Rf = 0.21, product Rf = 0.14), the reaction

131

mixture was poured into water (10 ml) and extracted with ethyl acetate (3 x 25 ml). The

132

extracts were combined and washed with water (30 ml) and sodium hydroxide solution (2 x

133

20 ml, 1 N), dried over anhydrous sodium sulfate and concentrated under reduced pressure.

134

The 0.84 g crude product was purified by column chromatography using n-hexane and EtOAc

135

(9:1, v/v) as an eluent, to afford purified 3-(1-naphthalenyloxy)-1-pheny-l-propanol (4, 0.58 g,

136

70.2%).

137

1

138

(m, 2H), 7.47 – 7.42 (m, 3H), 7.41 – 7.35 (m, 3H), 7.31 (t, J = 7.3 Hz, 1H), 6.81 (d, J = 7.6

139

Hz, 1H), 5.14 (dd, J = 8.2, 4.9 Hz, 1H), 4.38 – 4.33 (m, 1H), 4.23 – 4.18 (m, 1H), 2.46 – 2.39

140

(m, 1H), 2.37 – 2.30 (m, 1H). 13C NMR (CDCl3, 151 MHz): δ(ppm): 154.57, 144.30, 134.63,

141

128.74, 127.84, 127.67, 126.54, 126.00, 125.96, 125.71, 125.40, 121.96, 120.55, 104.93,

142

72.28, 65.51, 38.65. HRMS: calc. [M+Na]+ 301.1199, found [M+Na]+ 301.1204.

Ac ce p

te

d

M

an

124

H NMR (CDCl3, 600 MHz): δ(ppm): 8.29 – 8.24 (m, 1H), 7.85 – 7.80 (m, 1H), 7.54 – 7.48

143 144

2.5.

Synthesis of 4-phenyl-2H,3H,4H-naphtho[1,2-b]pyran (6)

145

To a solution of 4 (0.54 g, 1.9 mmol) in CH2Cl2 (8 ml) triethylamine (0.29 g, 2.9 mmol) and

146

4-dimethylaminopyridine (DMAP) (1.02 mg, 8.2 μmol) were added under nitrogen

147

atmosphere. The reaction mixture was cooled to 0 °C. Methanesulfonyl chloride (0.29 g, 2.5 5 Page 5 of 18

mmol) was added dropwise to the reaction mixture and stirred at 0°C for 2 h. The reaction

149

mixture was stirred at room temperature for additional 21 hours. After completion (checked

150

by TLC using n-hexane and EtOAc (9:1, v/v), starting material Rf = 0.12, product Rf = 0.64),

151

the reaction mixture was added to CH2Cl2 (30 ml) and extracted with 0.5 M HCl (2 x 20 ml),

152

saturated sodium bicarbonate solution (2 x 20 ml), washed with water (2 x 20 ml), dried over

153

anhydrous sodium sulfate and concentrated under reduced pressure. The 0.65 g crude product

154

was purified by column chromatography using n-hexane and dichloromethane (9:1, v/v) as an

155

eluent, to afford purified 4-phenyl-2H,3H,4H-naphtho[1,2-b]pyran (6, 0.34 g, 67.3%).

156

HRMS: calc. [M+H]+ 261.1274, found [M+H]+ 261.1282. NMR spectra are shown in

157

Supplementary Materials, 1H and 13C chemical shifts are reported in Table 1.

us

cr

ip t

148

158 3.

Results and discussion

160

3.1.

Detection of by-product

161

The synthesis of dapoxetine usually follows the process route shown in Scheme 1. The

162

advantage of this procedure over other methods, is that it also offers the possibility to easily

163

prepare N-demethylated derivatives, the major metabolites of the parent compound [17].

164

Using methylamine or ammonia in the last step will result in the active metabolites

165

desmethyldapoxetine or didesmethyldapoxetine, respectively. Our aim was to isolate (for the

166

first time) the mesylate intermediate (5) for further synthetic purposes. Quenching the

167

mesylation and allowing the mixture to warm up to room temperature resulted in a less polar

168

product compared to the starting material (4) instead of the expected mesylate. The unknown

169

product was isolated by column chromatography and characterized by mass spectrometry and

170

various NMR techniques for structure identification.

171

Ac ce p

te

d

M

an

159

172

3.2.

Structure elucidation of the by-product

173

Following the isolation, accurate mass measurement was performed to determine the

174

elemental compositions of the by-product. High-resolution positive ion mode ESI-MS

175

spectrum of the unknown revealed [M+H]+ at m/z 261.1282 suggesting a molecular formula

176

[M] of C19H16O. The error between observed mass and theoretical mass of [M+H]+ was below

177

3 ppm. This formula also coincide with the main product ion of dapoxetine, indicating a

178

neutral loss of the dimethylamine moiety [M]+ [18, 19]. The MS data clearly show that

179

position 4 is crucial in the new compound, however it can not be excluded that the formula of

180

C19H17O [M]+ arose from the in-source dissociation of the methylsulfonyl moiety.

6 Page 6 of 18

In order to unequivocally prove the lack of the methylsulfonyl group and to elucidate the

182

structure of the by-product, NMR experiments were applied.

183

The 1H NMR spectrum of the isolated compound confirmed the lack of the methylsulfonyl

184

moiety as methyl singlet was not detected in the aliphatic region. The chemical shift of the

185

aliphatic methine hydrogen decreased by 0.84 ppm, presumably due to the loss of the

186

electron-withdrawing hydroxyl group. Instead of the twelve aromatic protons expected for

187

compound 5 only eleven were found, suggesting that the aromatic part is also involved in the

188

structural change.

189

The Attached Proton Test (APT) NMR experiment is a common way to assign CH

190

multiplicities in 13C NMR spectra as it provides the information on all sorts of carbons within

191

one experiment. The APT spectrum revealed an extra quaternary carbon at 117.78 ppm and

192

the lack of an aromatic CH carbon. All these spectral changes suggested a cyclization in

193

which a new carbon-carbon bond was formed between the alicyclic C-4 and the aromatic C-5

194

carbons. The hypothesized ring-closure was confirmed by HMBC experiment.

195

Figure 2 shows the key long-range correlations between the aliphatic H-3 methylene protons

196

and the aromatic C-5 carbon (3JC,H) as well as the aliphatic H-4 methine resonance and the

197

aromatic C-5 (2JC,H) and C-14 carbon (3JC,H) confirming the formation of a new carbon-

198

carbon bond. The complete resonance assignment of the by-product (see Table 1.)

199

unequivocally proved the formation of the tricyclic 4-phenyl-2H,3H,4H-naphtho[1,2-b]pyran

200

structure.

Ac ce p

201

te

d

M

an

us

cr

ip t

181

202

3.3.

Formation of the new by-product

203

The in situ prepared intermediate, phenyl-propanediol-naphthyl ether mesylate plays a critical

204

role in the formation of the tricyclic naphtopyrane derivative. At low temperature the rate of

205

the by-product formation is low, however the rate increases with rising temperature. The

206

preparation of fused aromatic pyran derivatives are often accomplished by various transition

207

or noble metal catalysts' assisted Friedel-Crafts reactions under vigorous reaction conditions

208

[20]. The mild reaction conditions of the cyclization process in our case can obviously be

209

explained with the chemical properties of compound 5. In the facile and effective ring closure

210

of phenyl-propanediol-naphthyl ether mesylate the lone electron pair of the ethereal oxygen is

211

crucial (Fig. 3).

212

The conjugation between the electron pairs of the ethereal oxygen and the naphthalene moiety

213

increases the electron density of the aromatic system, thereby increasing its nucleophilic 7 Page 7 of 18

reactivity. Concurrently, the electrophilicity of the benzyl methylene increases due to the

215

adjacent phenyl ring and the strongly polarized mesyl group, facilitating the direct interaction

216

with the naphthalene and the splitting of the mesylate. The electrophilic aromatic substitution

217

process results in a cyclization product in which the developed positive charge can not only

218

lead to a temporary high-energy carbocation intermediate, but forming an oxonium

219

intermediate and thereby opening the possibility of a relatively low energy transition for the

220

cyclization. The pyranium intermediate rearomatizes in the last step and yields the neutral 4-

221

phenyl-naphtopyran derivative. This proposed mechanism was further supported by

222

performing the same reaction using optically active alcohol as the starting material. The

223

isolated product was proved to be racemic by circular dichroism measurements corroborating

224

the proposed mechanism in Figure 3. The new by-product is not only generated in the course

225

of the isolation of the mesylate. This impurity is always present in raw dapoxetine batches as

226

the ring closure competes with the nucleophilic substitution especially at room temperature.

an

us

cr

ip t

214

227 3.4.

HPLC-DAD analysis of the new process-related impurity

229

HPLC is certainly the most popular method in impurity profiling of active pharmaceutical

230

ingredients (APIs). Recent advances in this technology allow fast and robust separation and

231

quantification of API impurities often hyphenated with sensitive and selective spectral

232

methods. The newly isolated and identified 4-phenyl-2H,3H,4H-naphtho[1,2-b]pyran is a

233

potential process-related impurity of Dpx. As Dpx is not yet official in any pharmacopoeia,

234

impurity profiling of the API is rather incomplete. There is a single literature describing an

235

HPLC method for the separation of process-related impurities of Dpx (compound 4 and 5)

236

[16]. To promote the impurity profiling of Dpx, a gradient HPLC-UV method was developed

237

for the separation of the new tricyclic compound (6) and the known impurity of 4 and

238

dapoxetine. Figure 4 shows the optimized separation of the impurities (for details see section

239

2.1)

240

The performance of the developed method was characterized as follows. Calibration curves

241

were constructed with 7 concentrations in the range of 1 μg/ml through 1000 μg/ml. Each

242

concentration was injected three times. The regression was calculated by the method of least

243

squares. The LOD of the compounds were determined as the concentration yielding a signal

244

three times the noise of the baseline, while the LOQ as ten times the noise. Table 2

245

summarizes the linearity and the LOD and LOQ values of the three compounds.

Ac ce p

te

d

M

228

246 247

4.

Conclusion 8 Page 8 of 18

248

A novel by-product has been identified in the synthetic pathway of dapoxetine. The new

249

compound was isolated, identified, and characterized using high-resolution MS and NMR

250

techniques. The 1H and

251

were completely assigned. Reaction mechanism for the formation of the new impurity was

252

proposed and also confirmed by synthesis. To advance the impurity profiling of dapoxetine, a

253

reversed-phase gradient HPLC method was also developed.

C NMR resonances of the new impurity, 4-phenyl-naphtopyran

ip t

13

254 255

5.

256

The financial support from OTKA PD109373 is highly appreciated. Sz. Béni thanks the

257

Hungarian Academy of Sciences for the financial support under the János Bolyai Research

258

Scholarship.

us

cr

Acknowledgment

an

259 2606. References

[1] N.B. Modi, M.J. Dresser, M. Simon, D. Lin, D. Desai, S. Gupta, Single- and multiple-dose

262

pharmacokinetics of dapoxetine hydrochloride, a novel agent for the treatment of premature

263

ejaculation, J. Clin. Pharm. 46 (2006) 301-309.

264

[2] D.W. Robertson, D.C. Thompson, D.T. Wong, 1-phenyl-3-naphthalenyloxypropanamines and

265

their use as selective serotonin reuptake inhibitors, Eli Lilly and Co., 1992, US5135947A.

266

[3] Q.-h. Jiang, Y.-x. Xu, Y. Yang, H.-p. Mu, R. Wan, The progress in the synthetic methods of

267

dapoxetine, Chinese J. Med. Chem. 23 (2013) 417-421.

268

[4] P. You, J. Qiu, E. Su, D. Wei, Carica papaya Lipase Catalysed Resolution of β-Amino Esters for the

269

Highly Enantioselective Synthesis of (S)-Dapoxetine, Eur. J. Org. Chem. 2013 (2013) 557-565.

270

[5] K. Lin, Process for synthesis of Dapoxetine from trans-cinnamaldehyde and N-Cbz-

271

hydroxylamine, in, Hunan Ouya Biological Co., Ltd., Peop. Rep. China . 2013, pp. 13.

272

[6] G.L. Khatik, R. Sharma, V. Kumar, M. Chouhan, V.A. Nair, Stereoselective synthesis of (S)-

273

dapoxetine: A chiral auxiliary mediated approach, Tetrahedron Lett. 54 (2013) 5991-5993.

274

[7] G. Neumajer, T. Sohajda, A. Darcsi, G. Tóth, L. Szente, B. Noszál, S. Béni, Chiral recognition of

275

dapoxetine

276

electrophoresis method, J. Pharm. Biomed. Anal. 62 (2012) 42-47.

277

[8] R.K. Rapolu, V.V.N. Kali, V.P. Raju, G.M. Reddy, J. Pasha, N.R. Nevuluri, R. Bandichhor, S.

278

Oruganti, Short enantioselective routes to (S)-Dapoxetine, Chem. Biol. Interface, 3 (2013) 50-60.

Ac ce p

te

d

M

261

enantiomers

with

methylated-gamma-cyclodextrin:

A

validated

capillary

9 Page 9 of 18

[9] K.P. Mohan Rao Dodda, Jithender Aadepu, Solid dapoxetine, in, 2013.

280

[10] M. Sasikumar, M.D. Nikalje, Simple and efficient synthesis of (S)-dapoxetine, Synth. Commun.

281

42 (2012) 3061-3067.

282

[11] S. Rakesh, S.M.A. Hussain, A. Roy, A method for preparing (S)-(+)-N,N-dimethyl-3-(naphthalen-

283

1-yloxy)-1-phenylpropan-1-amine or its salt and intermediate thereof, in, R L Fine Chem, India .

284

2012, pp. 43.

285

[12] S.P.R. Nalla, D.V.K.R. Dandu, A process for the preparation of S-dapoxetine hydrochloride, in,

286

India. 2012, pp. 51.

287

[13] A.M. Dave, D.J. Patel, R. Kumar, S.D. Dwivedi, A process for preparing (+)-dapoxetine and its

288

salts, in, Cadila Healthcare Limited, India . 2008, pp. 16.

289

[14] P. Chen, G. Zhang, F. Yu, Process for preparation of dapoxetine hydrochloride, in, Shanghai

290

Maryao Chemical Technique Limited Company, Peop. Rep. China . 2006, pp. 12.

291

[15] C.A. Alt, R.L. Robey, M.E.E. Van, Intermediates to 1-phenyl-3-naphthalenyloxy-propanamines,

292

Eli Lilly and Co., 1994, US5292962A

293

[16] T.A. Rohith, S., Development and validation of high performance liquid chromatography

294

method for the determination of process related impurities in dapoxetine hydrochloride., Internat.

295

J. Res. Pharm. Chem. 3 (2013) 74-82.

296

[17] C.L. Hamilton, J.D. Cornpropst, Determination of dapoxetine, an investigational agent with the

297

potential for treating depression, and its mono- and di-desmethyl metabolites in human plasma

298

using column-switching high-performance liquid chromatography, J. Chromatogr. B, 612 (1993)

299

253-261.

300

[18] L. Li, M.-Y. Low, X. Ge, B.C. Bloodworth, H.-L. Koh, Isolation and structural elucidation of

301

dapoxetine as an adulterant in a health supplement used for sexual performance enhancement, J.

302

Pharm. Biomed. Anal. 50 (2009) 724-728.

303

[19] T.K. Kim, I.S. Kim, S.H. Hong, Y.K. Choi, H. Kim, H.H. Yoo, Determination of dapoxetine in rat

304

plasma by ultra-performance liquid chromatography–tandem mass spectrometry, J. Chromatogr.

305

B, 926 (2013) 42-46.

306

[20] V.C. Pandurang, A.D. Dattatray, A. Sudalai, Process for the production of 4-substituted

307

chromanes via gold catalysis, in, WO 2013/088455, 2013.

Ac ce p

te

d

M

an

us

cr

ip t

279

308 309 10 Page 10 of 18

310 311

Figure captions

312

Figure 1. Synthetic scheme of dapoxetine involving in situ mesylation in the last step

313 Figure 2. Inset of the 1H–13C gHMBCAD spectrum of 4-phenyl-2H,3H,4H-naphtho[1,2-

315

b]pyran showing the key long-range correlations for complete structure elucidation

ip t

314

cr

316

Figure 3. The proposed reaction mechanism for the formation of 4-phenyl-2H,3H,4H-

318

naphtho[1,2-b]pyran during dapoxetine synthesis

us

317 319

Figure 4. Reversed-phase HPLC-UV chromatograms of dapoxetine (Rt = 2.67 min) and the

321

impurities: 3-(1-naphthalenyloxy)-1-pheny-l-propanol (4) (Rt = 8.20 min) and 4-phenyl-

322

2H,3H,4H-naphtho[1,2-b]pyran (6) (Rt = 9.45 min) at 0.5% (top) and 0.1% (bottom)

323

concentration levels recorded at 310 nm..

324

Tables

325

Table 1. The numbering and the complete 1H and

326

product of dapoxetin. The 1H chemical shifts were referenced to TMS (0.00 ppm) while

327

chemical shifts were referenced to the applied NMR solvent CDCl3 (77.16 ppm).

329 330

M

d

C resonance assignment of the new by13

te

13

C

Ac ce p

328

an

320

Table 2. The linearity and the detection and quantitation limit of the developed LC method.

11 Page 11 of 18

2 3

O1

16 15 18

16

4

7

9

5 14 13

17

10

12 11

ip t

330 331

8

6

17

13 APT H, δ (ppm, multiplicity) C, δ (ppm) CH2 4.39-4.35 (m, 1H) 63.95 CH2 4.35-4.32 (m, 1H) 63.95 CH2 3 2.47-2.41 (m, 1H) 31.97 CH2 31.97 2.18-2.12 (m, 1H) CH 4 4.30 (t, J = 6.1 Hz, 1H) 41.05 C 5 117.78 C 6 150.50 C 7 125.37 CH 8 8.28 – 8.19 (m, 1H) 121.85 CH 9 7.50-7.44 (m, 1H) 125.44 CH 10 7.50-7.44 (m, 1H) 126.15 CH 11 7.78 – 7.71 (m, 1H) 127.57 C 12 133.61 CH 13 7.28 (d, J = 8.5 Hz, 1H) 119.79 CH 14 6.95 (d, J = 8.5 Hz, 1H) 128.45 C 15 146.04 2CH 16 7.15 (d, J = 7.2 Hz, 2H) 128.90 2CH 17 7.29 (t, J = 7.4 Hz, 2H) 128.57 CH 18 7.22 (t, J = 7.2 Hz, 1H) 126.58 1 13 H and C assignments: d, doublet; t, triplet; m, multiplet; J: coupling constants given in Hertz.

1

Ac ce p

332 333 334 335

te

d

M

an

us

cr

Position 2

12 Page 12 of 18

335 Dpx y = 0.2957x + 1.306 0.9998 0.51 1.7

compound 4 y = 0.4239x + 1.637 0.9999 0.47 1.56

compound 6 y = 0.5844x + 2.551 0.9997 0.44 1.46

ip t

regression determination coeff. LOD (! g/ml) LOQ (! g/ml)

Ac ce p

te

d

M

an

us

cr

336 337

13 Page 13 of 18

us

cr

i

*Graphical Abstract

O

MsCl Et3N

S

O

O

THF

ce pt

ed

0-5°C

Ac

O

CH3

M an

O

OH

(CH3)2NH

N

CH3

O

THF 0-5°C

Dapoxetine O

SEAr

SN2

H3C

-O S

CH3 O

rt °C !!! O

Dapoxetine impurity Page 14 of 18

Ac

ce

pt

ed

M

an

us

cr

i

Figure 1.

Page 15 of 18

cr Ac

ce

pt

ed

M

an

us

Figure 2.

Page 16 of 18

Ac

ce

pt

ed

M

an

us

cr

i

Figure 3.

Page 17 of 18

Ac

ce

pt

ed

M

an

us

cr

i

Figure 4.

Page 18 of 18

Structure elucidation of a process-related impurity of dapoxetine.

Unknown by-product associated with the synthesis of dapoxetine was isolated. The structure elucidation of this new compound using accurate mass data a...
2MB Sizes 3 Downloads 3 Views