Accepted Manuscript Title: Towards a real time release approach for manufacturing tablets using NIR spectroscopy Author: Aude Pestieau Fabrice Krier Gr´egory Thoorens Ana¨ıs Dupont Pierre-Franc¸ois Chavez Eric Ziemons Philippe Hubert Brigitte Evrard PII: DOI: Reference:

S0731-7085(14)00232-5 http://dx.doi.org/doi:10.1016/j.jpba.2014.05.002 PBA 9565

To appear in:

Journal of Pharmaceutical and Biomedical Analysis

Received date: Revised date: Accepted date:

24-2-2014 28-4-2014 1-5-2014

Please cite this article as: A. Pestieau, F. Krier, G. Thoorens, A. Dupont, P.-F. Chavez, E. Ziemons, P. Hubert, B. Evrard, Towards a real time release approach for manufacturing tablets using NIR spectroscopy, Journal of Pharmaceutical and Biomedical Analysis (2014), http://dx.doi.org/10.1016/j.jpba.2014.05.002 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

Towards a real time release approach for manufacturing tablets using NIR spectroscopy

2 Aude Pestieau1, Fabrice Krier1, Grégory Thoorens1, Anaïs Dupont1, Pierre-François Chavez2,

4

Eric Ziemons2, Philippe Hubert2, Brigitte Evrard1

ip t

3

5 6

1

7

of Liège, 4000 Liège, Belgium

8

2

9

Liège, 4000 Liège, Belgium

10

Corresponding author

cr

Laboratory of Pharmaceutical Technology, Department of Pharmacy, C.I.R.M., University

an

us

Laboratory of Analytical Chemistry, Department of Pharmacy, C.I.R.M., University of

Aude Pestieau

12

[email protected]

13

+3243664306

14

CHU, Tour 4, 2nd floor, Laboratory of Pharmaceutical Technology, Department of Pharmacy,

15

University of Liège, Avenue de l'Hôpital, 1, 4000 Liège, Belgium

17

d

te

Ac ce p

16

M

11

Highlights

18

‐ 

We used NIR Spectroscopy to control in‐line blends uniformity. 

19

‐ 

We used NIR Spectroscopy to evaluate the conformity of paracetamol tablets. 

20

‐ 

Mixing time of blend could be reduced because NIR showed when it was homogenous. 

21

‐ 

Tablet NIR analyses successfully allowed the prediction of their conformity.  

22

‐ 

The benefit of NIR methods is significant for reducing batch release time. 

23 1 Page 1 of 23

24

25

1. Introduction Conventional pharmaceutical manufacturing is generally accomplished using batch

27

processing with laboratory testing conducted on collected samples to evaluate quality. This

28

conventional approach has been successful in providing quality drug products to the public.

29

However, quality controls on process materials and on finished products are time-consuming

30

and often require a lot of sample preparation steps and laboratory work. For a few years,

31

significant opportunities have existed for improving pharmaceutical development,

32

manufacturing and quality assurance through innovation in product and process development,

33

process analysis, and process control. For example, the publication of the Process Analytical

34

Technology (PAT) initiative [1] by the Food and Drug Administration (FDA)has increased

35

the interest for PAT tools in the pharmaceutical industry. One of the principles described in

36

the guidance document is Real Time Release (RTR) which can be defined as the ability to

37

evaluate and ensure the acceptable quality of in-process and/or final product based on process

38

data [2]. The combined process measurements and other test data gathered during the

39

manufacturing process can serve as the basis for RTR of the final product and would

40

demonstrate that each batch conforms to established regulatory quality attributes. According

41

to the European Medicines Agency (EMA), “Real Time Release Testing (RTRT) will, in

42

general, comprise a combination of process controls which may utilise PAT tools e.g. NIR

43

and Raman spectroscopy (usually in combination with multivariate analysis), together with

44

the control of relevant material attributes.”[3]. For example, a well-known application of this

45

concept is the use of NIRS to control the blend uniformity with an in-line measurement. This

46

type of application is very useful in the context of RTR of the product by reducing the batch

47

release time [4]. Moreover, the blend homogeneity control is a crucial step to ensure active

Ac ce p

te

d

M

an

us

cr

ip t

26

2 Page 2 of 23

content uniformity in the end product, especially in a direct compression process, because it is

49

the only step before compression. Indeed, the present manufacturing process time compared

50

to the time spent on quality testing after manufacturing is very low, so RTR offers some

51

significant benefits to the manufacturer [5].

ip t

48

52

To perform this implementation, the use of analytical techniques able to provide accurate results in a simple and a rapid way is required. Due to its non-destructive nature and its

54

immediate (real time) delivery of results without sample preparation, NIRS is a suitable

55

analytical technique for this goal. This is probably why the European Pharmacopeia has

56

decided to add a monograph on this technique in the latest published version (version 8.0.)

57

[6].

us

an

However, after the development of the NIRS method, another major step was validation

M

58

cr

53

enabling its routine use. Validation is based on guidelines of the International Conference on

60

Harmonisation (ICH) Q2 (R1) [7] and is a crucial and mandatory step in the lifecycle of an

61

analytical method. This step is needed to prove that the spectroscopic analytical method is

62

suitable for its intended use and consequently, to show the reliability of the results obtained

63

within well-defined limits. It integrates all the useful required validation criteria such as

64

accuracy, trueness, precision, limits of quantification, range and linearity. The approach based

65

on the accuracy profile makes the visual and reliable representation of the future

66

performances of the analytical method possible and thus, enables better risk management [8].

67

In this manner, it also complies with the ICH Q9 regulatory documents [9].

te

Ac ce p

68

d

59

In the pharmaceutical industry, NIRS combined with chemometrics have already been

69

applied to acquire information on a particular quality attribute or on multiple predicates.

70

There are many examples in the literature of acquiring information on a particular quality

71

attribute such as the active content [5, 8], the blend homogeneity [10], the coating level [11,

72

12], the moisture content [13], the polymorphic transformations [14], the particle size of

3 Page 3 of 23

powders [15], the flow property of powder[16], the tablet mechanical strength [17] and the

74

dissolution profiles [18].However, for all these applications, only few analytical methods have

75

been validated. In fact, the validation step was only performed on the active content

76

determination in tablets [5] or in pellets [8] and on the moisture content determination in

77

pellets [13]. A number of successful combinations of two or three qualitative and/or

78

quantitative simultaneous determinations are already described. Simultaneous determination

79

of content uniformity and tablet hardness [19] or blend uniformity, content uniformity and

80

coating thickness [4] or tablet hardness, content uniformity and dissolution test [20] on intact

81

tablets can be found. However in these studies, only the NIR method for the determination of

82

the API content was validated. Regarding oral dosage forms, the literature also shows the

83

paracetamol determination in low-dose pharmaceutical syrup [21]or in matrix carrier

84

produced by hot-melt extrusion [22]. Of course, this technique can be applied to other dosage

85

forms that are not intended for oral administration such as the in-line monitoring of the

86

implant manufacturing process [23] and the determination of API and preservatives in a gel

87

[24].

89 90

cr

us

an

M

d

te

In the present study, tablets for oral administration were selected. At first NIRS has been

Ac ce p

88

ip t

73

used to control the blend uniformity. It was an in-line measurement. Following the manufacture of tablets, we attempted to develop and validate NIR methods

91

that can replace the conventional techniques usually used to test tablets following their

92

manufacture, ie. the European Pharmacopeia tests for content uniformity, hardness,

93

disintegration time and friability. To the best of our knowledge, no literature describes the use

94

of NIR to replace four Pharmacopeia tests simultaneously.

4 Page 4 of 23

95

2. Materials and methods 2.1. Chemicals

97

Paracetamol (CompapTM PVP3) was provided by Covidien (Mallinckrodt, USA).

ip t

96

Microcrystalline cellulose (Avicel®PH-102) was provided by FMCBioPolymer (Cork,

99

Ireland). Sodium starch glycolate (Glycolys®) was supplied by Roquette (Lestrem, France).

100

cr

98

Magnesium stearate was obtained from Fagron (Waregem, Belgium).

All solvents used in the reference methods were of analytical grade. Methanol was

102

purchased from J.T. Baker® (Deventer, Netherlands). Water was purified by a Millipore®

103

system (18.2 MΩ/cm resistivity, Milli-Q) before filtration through a 0.22 µm Millipore

104

Millipak® – 40 disposable filter units (Millipore Corporation, USA).

M

an

us

101

2.2. Tablets manufacturing

106

The tablet formulation was developed for direct compression and based on paracetamol as

107

API. This API was previously wet granulated with 3% of polyvinylpyrrolidone and therefore

108

easily compressible. The excipients that have been added to this Paracetamol CompapTM

109

PVP3 are a binder (microcrystalline cellulose), a super-disintegrant (sodium starch glycolate)

110

and a lubricant (magnesium stearate). This formulation represents a typical formulation for

111

the manufacture of tablets by direct compression and provides optimum flow properties.

te

Ac ce p

112

d

105

Paracetamol tablets were manufactured by direct compression with an eccentric press

113

(AC27, GEA-Courtoy, Halle, Belgium). Blends were mixed in a high shear mixer Gral-

114

10®(GEA-Colette, Wommelgem, Belgium). The process bowl has a volume of 10 l. The

115

blend load was 2.4 kg. The mixing was performed for 400 s without magnesium stearate at

116

200 rpm. The lubricant was then added and mixed during 1 min. Flat face bevel edge tablets

117

were obtained using round punches with a diameter of 10 mm. Targeted tablet weight was

118

fixed at 350 mg. 5 Page 5 of 23

On the one hand, three different active pharmaceutical ingredient concentrations were

120

manufactured: 80, 100 and 120 % of a predetermined dosage (242.5 mg of paracetamol) at a

121

compaction pressure of 45 kg/cm2. To obtain these different concentrations, the weight of the

122

API was increased or decreased relative to the target concentration. As can be seen in Table 1,

123

the quantity of microcrystalline cellulose and sodium starch glycolate were adapted to

124

maintain a tablet weight of 350 mg while the quantity of magnesium stearate remained

125

constant (1 % w/w). Indeed, during an industrial production of tablets, uniformity of mass is a

126

parameter which is very often controlled. In this way a de-mixing of paracetamol and

127

excipients during tableting should be simulated.

130

cr

us

an

129

On the other hand, two additional compaction pressures were used for tablets with an API concentration of 100 %: 25 and 65 kg/cm2.

M

128

ip t

119

For the calibration and external validation, one batch of tablets was manufactured each day during three days (D1, D2 and D3) for each concentration (C80, C100, C120) and for each

132

compaction pressure (CP25, CP45, CP65).One batch consisted of approximately 6,000 tablets.

te

d

131

2.3. FT-NIR equipment

134

Intact tablets were analyzed by transmission mode with a multipurpose analyzer Fourier

Ac ce p

133

135

transform near infrared spectrometer (MPA, Bruker Optics, Ettlingen, Germany). The spectra

136

were collected with the Opus software 6.5 (MPA, Bruker Optics, Ettlingen, Germany). Each

137

spectrum was an average of 32 scans and the resolution was 8 cm-1 over the range from 3600

138

to 14000 cm-1.For in-line monitoring of the blending process, the NIR spectrometer was

139

equipped with a NIR reflectance probe (Series 400 Diffuse Reflectance probe, Precision

140

Sensing Devices, Massachusetts, USA) interfaced with the mixing bowl. Each spectrum was

141

an average of 4 scans and the resolution was 16 cm-1 over the range from 12500 to 4000 cm-1.

142

The time required for a NIR measurement was 1.7 sec and the time interval between measures

143

was 1 sec. 6 Page 6 of 23

144

2.4. Reference methods

145

A conventional evaluation blend uniformity method was performed as a reference [25].

ip t

146

2.4.1. Powder blend uniformity

This method involved blending for a pre-determined length of time (400 s), stopping the

148

blender, and manually withdrawing a powder blend sample representative of a unit dose from

149

the bin with a single compartment end-sampling thief probe. Ten locations were selected from

150

two depths along the axis of the bowl. The samples were then analysed using a USP high

151

performance liquid chromatography (HPLC) method.

us

This HPLC method uses a GraceSmart® steel column 25 cm long x 4,6 mm ID RP 18 with

an

152

cr

147

particles of 5 µm, a mobile phase consisting of methanol-water (3:1, v/v) flowing at 1.5

154

ml/min, an injected volume of 10 µl, a chromatographic run time of 6 min and a detection

155

wavelength of 243 nm. The HPLC apparatus used was an Agilent 1100® (Santa Clara, United

156

State).

d

M

153

Before injection, each sample was weighed, dissolved in 20.0 ml of mobile phase, and

158

sonicated for 5 min. An aliquot of 0.5 ml was then diluted to 200.0 ml with the mobile phase.

159

This solution was filtered through a 0.22 µm polyvinylidene fluoride (PDVF)filter. A standard

160

was prepared from pure paracetamol in the same dilution solvent.

Ac ce p

161

te

157

We considered samples to be uniform if the drug concentration of each individual sample

162

was within 10% of the average concentration and the relative standard deviation (RSD) was

163

less than 5%.

164

2.4.2. Content uniformity

165

The same USP HPLC method as described above was used as a reference method to

166

determine tablet content uniformity. Each tablet was weighed, dissolved in 100 ml of mobile

167

phase, sonicated for 15 min and diluted to 250 ml with the same solvent. An aliquot of 1 ml 7 Page 7 of 23

168

was then diluted to 100 ml with the mobile phase. This solution was filtered through a 0.22

169

µm PDVF filter before injection.

2.4.3. Tablet hardness

171

Tablet hardness was determined using a Sotax HT1®(Allschwil, Switzerland). This

ip t

170

apparatus consists of two jaws facing each other, one of which moves towards the other. The

173

flat surfaces of the jaws are perpendicular to the direction of movement and the tablet was

174

placed horizontally between the jaws [26].

us

cr

172

2.4.4. Disintegration time

176

For the determination of tablet disintegration time, a Sotax DT3® (Allschwil,

177

Switzerland) was used. This apparatus consists of a basket-rack assembly, a 1 liter, low-form

178

beaker for the immersion fluid, a thermostatic arrangement for heating the fluid at 37 ± 2 °C

179

and a device for raising and lowering the basket in the immersion fluid at a constant

180

frequency rate. The basket-rack assembly moves vertically along its axis. One dosage unit

181

was placed in one of the 6 tubes of the basket and time where the dosage unit has

182

disintegrated completely was noted. This method corresponds to the reference [27].

184

M

d

te

Ac ce p

183

an

175

2.4.5. Tablet friability

For tablet friability, the method used corresponds to the reference [28]with a

185

Friabilitator Sotax F1/2® (Allschwil, Switzerland). This equipment consists of a drum made of

186

transparent synthetic polymer. The tablets are tumbled at each rotation of the drum by a

187

curved projection that extends from the middle of the drum to the outer wall. The drum is

188

attached to the horizontal axis of a device that rotates 100 times in 4 minutes (25 ± 1 rpm).

189

Thus, at each rotation the tablets roll or slide and fall onto the drum wall or onto each other. A

190

sample of whole tablets corresponding as near as possible to 6.5 g (19 tablets) was tested.

8 Page 8 of 23

These tablets were carefully dusted and then weighed prior to testing. At the end of the test,

192

the tablets were removed from the drum, dusted and then reweighed. If tablets that were

193

obviously cracked, cleaved, or broken were present in the tablet sample after tumbling, the

194

sample failed the test. A maximum loss of mass less than 1.0 % is considered acceptable for

195

most products.

ip t

191

2.5. Development of calibration models

us

197

cr

196

2.5.1. Powder blend uniformity

199

For in-line monitoring, the mixing bowl was perforated to allow the introduction of a

an

198

NIR fibre probe. This equipment was widely described by Bodson in [29].

201

The conformity test was used to follow the mixing kinetics [29]. The conformity test is an

202

easy method to test the deviation of measured NIR spectra. To set limits, samples of the final

203

product are needed as reference spectra which belong to at least one batch or one production

204

cycle. These reference spectra vary within the accepted range of specifications. The NIR

205

spectra of these samples reflect the different sample variations and give a confidence band in

206

the spectral range. To pass the conformity test, the spectrum of a new sample has to be within

207

this confidence band at each wavelength. The conformity tests were computed with Opus

208

software 6.5(Bruker Optics, Ettlingen, Germany) and the Conformity Index (CI) was

209

calculated using the following equation:

d

te

Ac ce p

210

M

200

Conformity Index = (Āref,i - Asamp,i) / σref,i[30]

211

Āref,i : Average of absorbance values of reference spectra at wavenumber i.

212

Asamp,i : Absorbance value of test sample spectrum at wavenumber i.

213

σref,i : Standard deviation of absorbance values of reference spectra at

214

wavenumber i. 9 Page 9 of 23

215

The last ten spectra of each blend were selected as reference spectra.

216

The selected algorithm added up all y-values above the CI limit and divided this sum by the

217

total number of data points within the frequency ranges selected [30].

219

2.5.2. Pharmacopeia tests

ip t

218

To build the NIR model for content uniformity, twenty-seven tablets were analysed.

Indeed, as mentioned above three levels of concentrations (C80, C100, C120) were produced

221

each day during three days (D1, D2D3) at a compaction pressure of 45 kg/cm2(CP45)and 3

222

tablets were measured per production (3 repetitions). To build the NIR models for tablet

223

hardness and disintegration time, twenty-seven tablets were also analysed. One level of

224

concentration (C100) was compressed each day during three days (D1, D2 D3) at three

225

compaction pressures (CP25, CP45, CP65) and 3 tablets were measured per production (3

226

repetitions).Theseproductionswerealso used for tabletfriability testing,butfor this model19

227

tabletswere analyzedper production. So, hundred seventy one tablets were analysed to build it.

228

For each validated model the same number of tablets as used for calibration was produced

230

us

an

M

d

te

and measured for the external validation.

Ac ce p

229

cr

220

In a first step, a PLS regression model was built for each test using calibration samples.

231

Pre-treatments for offset baseline correction, sample normalization and variable centering

232

were performed as Standard Normal Variate (SNV), Multiplicative Scatter Correction (MSC),

233

Mean Centering, derivatives, detrend and smoothing. Cross-validation based on random

234

subsets was carried out to select the optimal number of PLS factors for API concentrations,

235

tablet hardness and disintegration time tests. For tablet friability, a qualitative approach was

236

adopted by a KNN analysis, whichallowed us to detect non-conforming tablets.

237

All data were mean centred and the number of latent variables and of each PLS model was

238

selected based on Root Mean Square Error of Cross-Validation (RMSECV) versus latent

239

variable plots. 10 Page 10 of 23

240

The best models were chosen based on their RMSEP.

241

PLS models were built using PLS_Toolbox 7.0.3 (Wenatchee, WA, USA) running on Matlab R2013a (The Mathworks, Natick, MA, USA).

243

3. Results and discussion

ip t

242

3.1. Powder blend uniformity

245

To confirm blend uniformity of the final blends, the reference method described above

us

cr

244

was used. The spectral areas from 9033 to 8277 cm-1and from 7212 to 4242 cm-1,

247

corresponding to absorption bands of paracetamol, were selected to compute the conformity

248

tests. SNV was used as pre-processing. As the conformity curve represents the spectral

249

variability during the blending step, the blend can be assessed as homogeneous when the

250

curve reaches a stable level. In Figures 1, 2 and 3, green points are the reference spectra

251

corresponding to the end of the mixing process when the blend is expected to be

252

homogeneous, while blue points are sample spectra of the blend from the beginning to the end

253

of the mixing process. As can also be seen in these figures, the 80%, 100% and 120% blends

254

seem to be homogeneous before the end of the mixing time. This homogeneity has been

255

confirmed by thieved sampling after the blending process and HPLC analysis with the

256

reference method described above. The results obtained with this technique are shown in

257

Table 2. As can be seen in this table, the means of sample content uniformity were close to

258

the target value and the coefficient of variation was less than 2.5 % for all blends. These

259

results show that blends are effectively homogenous after 400 s of mixing. Since all the

260

conformity curves reach the final homogeneity state before the end of the process, it can be

261

concluded that the blends are homogeneous before the 400 s and therefore, this time could be

262

reduced.

Ac ce p

te

d

M

an

246

11 Page 11 of 23

3.2. NIR models for Pharmacopeia tests

264

As can be seen in Table 4, a spectral range was manually selected for the calibration of

265

each NIR model. In a general way, as seen in Figure 4, the region above 7500 cm-1(or 9000

266

cm-1) was selected because below this wavenumber, the detector signal becomes noisy.

267

ip t

263

This table also shows the spectral pre-treatment used, the number of PLS factors selected and the R2, RMSEC and RMSEPobtained for each test. In fact, the validation step using the

269

accuracy profile approach was performed for the content uniformity method and for the tablet

270

hardness method. Due to the uncertainty of the reference method for disintegration time

271

(subjective visual method), the validation step was based on the values of RMSEC and

272

RMSEP. Finally, regarding tablet friability, a qualitative approach was performed in order to

273

detect non-conforming tablets.

276 277

us

an

M

d

Table 4 shows that three PLS factors were selected for the NIR model with regard to the

te

275

3.2.1. Content uniformity

lowest value of RMSEC. It was at 0.415for all three factors. This NIR method was validated and the accuracy and risk profiles were evaluated. The

Ac ce p

274

cr

268

278

acceptance limits were set at ±10% in accordance with USP recommendations [31].As it can

279

be seen on Figure 5, the method was successfully validated for the entire dosage interval from

280

80% to 120%. Furthermore, the relative bias was very close to 0 % for each concentration

281

level and the distribution of the random errors was quite low too. In fact we can say that this

282

method presents excellent trueness and precision.

283 284

3.2.2. Tablet hardness As a reminder, three compaction pressures were used to build this model. In fact, these

285

compaction pressures were selected according to the mechanical constraints of the eccentric

286

press and the formulation. Indeed, a compaction pressure below 25 kg/cm2 gave weak tablets, 12 Page 12 of 23

which were difficult to handle and had very low hardness values. Conversely, a compaction

288

pressure above 65 kg/cm2 would produce excessive mechanical stress to the tablet press.

289

Regarding the third compaction pressure, it was the median value between these two

290

extremes, that is to say 45 kg/cm2. However, this compaction pressure provides the

291

appropriate hardness to this type of tablet for oral use (80 N – 120 N). Indeed, a lower

292

hardness would lead to tablets that are too friable and would not withstand the handling

293

required for their packaging. Conversely, a higher hardness would reduce disintegration time

294

and may decrease the bioavailability and the effectiveness of the drug.

cr

us

We can see in Figure 6 that the acceptance limits were set at ± 25% for this validation

an

295

ip t

287

method. The β-expectation tolerance limits are included in these acceptance limits for

297

hardness values between 60 N and 160 N. As explained above, these hardness values are in

298

agreement with the expected values for this type of tablet. It is reasonable to admit that tablets

299

having a hardness of less than 60 N or greater than 160 N will be considered as non-

300

conforming. In this case, it is no longer necessary to try to accurately quantify their hardness.

301

It can be concluded that, during routine use, this method will provide results with adequate

302

accuracy. The risk of finding future results outside the ±25% acceptance limit is below 5%,

303

which is the chosen maximum risk level (α-risk: 5%).

d

te

Ac ce p

304

M

296

3.2.3. Disintegration time

305

The PLS model chosen for tablet disintegration time required three factors. The NIR

306

model was tested by the calibration and the validation sets. Figure 7 shows NIR predictions

307

versus reference method results for these two sets. The calibration set is represented by the

308

red triangles and the validation set by the black points. The values of the RMSEC and the

309

RMSEP were 1.67 and 4.18 respectively. These results indicate the robustness and the global

310

accuracy of the NIR model. It is important to keep in mind that the reference method is a

311

visual subjective method. Indeed, the disintegration end-point is left to the judgment of the 13 Page 13 of 23

operator and is defined as “that state in which any residue of the unit, except fragments of

313

insoluble coating or capsule shell, remaining on the screen of the test apparatus or adhering

314

to the lower surface of the discs”[27]. Thus, it is difficult to obtain accurate and robust

315

quantitative results with this technique. However, the conformity of uncoated tablets for the

316

disintegration test is achieved when disintegration time is less than 15 min. In our case, all

317

tested tablets used to build the model disintegrated before one minute of testing was complete.

318

Thus, one can reasonably assume that the NIR model will easily detect non-conforming

319

tablets. To test this hypothesis, it would be necessary to have tablets with poor disintegration.

320

This seems to be difficult to obtain with the current formulation. Indeed, compaction pressure

321

could not be increased without exceeding the maximum mechanical stress of the tablet press

322

and tablets were uncoated.

M

an

us

cr

ip t

312

3.2.4. Tablet friability

324

For this test, according to [28], a sample of whole tablets corresponding as near as

d

323

possible to 6.5 g have to be tested. This engenders the analysis of 19 tablets in a single

326

friability test. Under these conditions, it seems to be difficult to do a quantitative analysis. The

327

classification for this test was based on KNN, a qualitative approach. To account for scaling

328

effects and offset effects, a MSC was applied as spectra pre-treatment.

Ac ce p

329

te

325

Figure 7 shows the results of the multivariate pattern recognition method KNN. With

330

this approach, it can be seen that the NIR method was able to classify tablets on two levels

331

(levels 1 and 2 in Fig.8). After comparison of these results with those of the reference method,

332

it appeared that these two levels correspond to samples that failed the friability test (in red)

333

and samples that passed it (in green). Thus this qualitative approach allows us to determine

334

whether or not tablets conform in terms of friability.

14 Page 14 of 23

335 336

4. Conclusions During this study, we showed that NIRS is an interesting tool as a RTR system. First, we developed a real-time, non-invasive, in-line qualitative blend uniformity method. The

338

homogeneity has been successfully confirmed by thieved sampling after the blending process

339

(400 s) and HPLC analyses. Thanks to the NIRS method it was shown that all the conformity

340

curves reach the final homogeneity state before the end of the process. In fact, with the use of

341

NIR for this application, an overview of the blend homogeneity over time is given, whereas

342

by the reference method, only specific results can be obtained. In our case, NIR results

343

showed that the blends are homogenous before the 400 s and therefore, this time could be

344

reduced.

cr

us

an

After that, we successfully developed and validated four off-line NIRS methods intended

M

345

ip t

337

to replace the conventional Pharmacopeia tests for content uniformity, tablet hardness,

347

disintegration time and tablet friability. Taking into account that the quality control of

348

finished products is time-consuming, this nondestructive method offers significant

349

advantages, especially in terms of batch release time. Thanks to the rapidity and the ease of

350

use of this method once developed, NIRS can be considered as a quality control technique and

351

a cost effective solution.

te

Ac ce p

352

d

346

353

Abbreviations

354

NIRS

Near-Infrared Spectroscopy

355

API

Active Pharmaceutical Ingredient

356

PLS

Partial Least Squares

357

RMSEC

Root Mean Squared Error of Calibration

358

RMSEP

Root Mean Squared Error of Prediction 15 Page 15 of 23

KNN

K-Nearest-Neighbors

360

PAT

Process Analytical Technology

361

FDA

Food and Drug Administration

362

RTR

Real Time Release

363

EMA

European Medicines Agency

364

RTRT

Real Time Release Testing

365

ICH

International Conference of Harmonisation

366

USP

United State Pharmacopeia

367

HPLC

High Performance Liquid Chromatography

368

PVDF

PolyVinyliDene Fluoride

369

RSD

Relative Standard Deviation

370

CI

Conformity Index

371

SNV

Standard Normal Variate

372

MSC

Multiplicative Scatter Correction

373

RMSECV

Root Mean Squared Error of Cross-Validation

375

cr us

an

M

d

te

Ac ce p

374

ip t

359

Acknowledgements

376

Thanks to SMB Technology, FMCBioPolymer and Roquette for supplying us with some

377

excipients.

378 379 380 381 382 383 384 385 386

References [1] FDA, Guidance for Industry PAT - A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance, (2004). [2] E.T.S. Skibsted, J.A. Westerhuis, A.K. Smilde, D.T. Witte, Journal of Pharmaceutical and Biomedical Analysis, Examples of NIR based real time release in tablet manufacturing, 43 (2007) 1297-1305. [3] EMA, Guideline on Real Time Release Testing, (2012). 16 Page 16 of 23

te

d

M

an

us

cr

ip t

[4] J.J. Moes, M.M. Ruijken, E. Gout, H.W. Frijlink, M.I. Ugwoke, International Journal of Pharmaceutics, Application of process analytical technology in tablet process development using NIR spectroscopy: Blend uniformity, content uniformity and coating thickness measurements, 357 (2008) 108-118. [5] C. Bodson, E. Rozet, E. Ziemons, B. Evrard, P. Hubert, L. Delattre, Journal of Pharmaceutical and Biomedical Analysis, Validation of manufacturing process of Diltiazem HCl tablets by NIR spectrophotometry (NIRS), 45 (2007) 356-361. [6] Monograph 2.2.40. Near-Infrared Spectroscopy, in: Eur.Ph. 8.0., 2014. [7] ICH, Topic Q2 (R1): Validation of Analytical Procedures: Text and Methodology, International Conference on Harmonization of Technical Requirements for registration of Pharmaceuticals for Human Use, (2005). [8] J. Mantanus, E. Ziémons, E. Rozet, B. Streel, R. Klinkenberg, B. Evrard, J. Rantanen, P. Hubert, Talanta, Building the quality into pellet manufacturing environment – Feasibility study and validation of an in-line quantitative near infrared (NIR) method, 83 (2010) 305-311. [9] ICH, Topic 9 : Quality risk management, International Conference on Harmonisation of Technical requirements for registration of pharmaceuticals for human use, (2005). [10] Y. Sulub, B. Wabuyele, P. Gargiulo, J. Pazdan, J. Cheney, J. Berry, A. Gupta, R. Shah, H. Wu, M. Khan, Journal of Pharmaceutical and Biomedical Analysis, Real-time on-line blend uniformity monitoring using near-infrared reflectance spectrometry: A noninvasive offline calibration approach, 49 (2009) 48-54. [11] M.-J. Lee, D.-Y. Seo, H.-E. Lee, I.-C. Wang, W.-S. Kim, M.-Y. Jeong, G.J. Choi, International Journal of Pharmaceutics, In line NIR quantification of film thickness on pharmaceutical pellets during a fluid bed coating process, 403 (2011) 66-72. [12] C.V. Möltgen, T. Puchert, J.C. Menezes, D. Lochmann, G. Reich, Talanta, A novel inline NIR spectroscopy application for the monitoring of tablet film coating in an industrial scale process, 92 (2012) 26-37. [13] J. Mantanus, E. Ziémons, P. Lebrun, E. Rozet, R. Klinkenberg, B. Streel, B. Evrard, P. Hubert, Analytica Chimica Acta, Moisture content determination of pharmaceutical pellets by near infrared spectroscopy: Method development and validation, 642 (2009) 186-192. [14] M. Blanco, M. Alcalá, J.M. González, E. Torras, Analytica Chimica Acta, Near infrared spectroscopy in the study of polymorphic transformations, 567 (2006) 262-268. [15] M. Blanco, A. Peguero, Talanta, An expeditious method for determining particle size distribution by near infrared spectroscopy: Comparison of PLS2 and ANN models, 77 (2008) 647-651. [16] X. He, X. Han, N. Ladyzhynsky, R. Deanne, Powder Technology, Assessing powder segregation potential by near infrared (NIR) spectroscopy and correlating segregation tendency to tabletting performance, 236 (2013) 85-99. [17] J.D. Kirsch, J.K. Drennen, Journal of Pharmaceutical and Biomedical Analysis, Nondestructive tablet hardness testing by near-infrared spectroscopy: a new and robust spectral best-fit algorithm, 19 (1999) 351-362. [18] Y. Hattori, M. Otsuka, Vibrational Spectroscopy, NIR spectroscopic study of the dissolution process in pharmaceutical tablets, 57 (2011) 275-281. [19] M. Blanco, M. Alcalá, Analytica Chimica Acta, Content uniformity and tablet hardness testing of intact pharmaceutical tablets by near infrared spectroscopy: A contribution to process analytical technologies, 557 (2006) 353-359. [20] M. Blanco, M. Alcalá, J.M. González, E. Torras, Journal of Pharmaceutical Sciences, A process analytical technology approach based on near infrared spectroscopy: Tablet hardness, content uniformity, and dissolution test measurements of intact tablets, 95 (2006) 2137-2144.

Ac ce p

387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434

17 Page 17 of 23

te

d

M

an

us

cr

ip t

[21] E. Ziémons, J. Mantanus, P. Lebrun, E. Rozet, B. Evrard, P. Hubert, Journal of Pharmaceutical and Biomedical Analysis, Acetaminophen determination in low-dose pharmaceutical syrup by NIR spectroscopy, 53 (2010) 510-516. [22] P.R. Wahl, D. Treffer, S. Mohr, E. Roblegg, G. Koscher, J.G. Khinast, International Journal of Pharmaceutics, Inline monitoring and a PAT strategy for pharmaceutical hot melt extrusion, 455 (2013) 159-168. [23] F. Krier, J. Mantanus, P.-Y. Sacré, P.-F. Chavez, J. Thiry, A. Pestieau, E. Rozet, E. Ziemons, P. Hubert, B. Evrard, International Journal of Pharmaceutics, PAT tools for the control of co-extrusion implants manufacturing process, 458 (2013) 15-24. [24] M. Blanco, M. Alcalá, M. Bautista, European Journal of Pharmaceutical Sciences, Pharmaceutical gel analysis by NIR spectroscopy: Determination of the active principle and low concentration of preservatives, 33 (2008) 409-414. [25] F.J. Muzzio, C.L. Goodridge, A. Alexander, P. Arratia, H. Yang, O. Sudah, G. Mergen, International Journal of Pharmaceutics, Sampling and characterization of pharmaceutical powders and granular blends, 250 (2003) 51-64. [26] Monograph 2.9.8. Resistance to crushing of tablets, in: Eur.Ph. 8.0., 2014. [27] Monograph 2.9.1. Desintegration of tablets ans capsules, in: Eur.Ph. 8.0., 2014. [28] Monograph 2.9.7. Friability of uncoated tablets, in: Eur.Ph. 8.0., 2014. [29] C. Bodson, W. Dewé, P. Hubert, L. Delattre, Journal of Pharmaceutical and Biomedical Analysis, Comparison of FT-NIR transmission and UV–vis spectrophotometry to follow the mixing kinetics and to assay low-dose tablets containing riboflavin, 41 (2006) 783-790. [30] J. Mantanus, E. Rozet, K. Van Butsele, C. De Bleye, A. Ceccato, B. Evrard, P. Hubert, E. Ziémons, Analytica Chimica Acta, Near infrared and Raman spectroscopy as Process Analytical Technology tools for the manufacturing of silicone-based drug reservoirs, 699 (2011) 96-106. [31] Acetominophen tablets monograph, in: USP 32, 2010.

Ac ce p

435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463

18 Page 18 of 23

8°80 % % 200 mg (57%) 120 mg (34%) 26,5 mg (8%) 3.5 mg (1%) 350 mg

Paracetamol + 3% PVP Microcrystalline cellulose Sodium starch glycolate Magnesium Stearate Total

100 %% 250 mg (71%) 79 mg (23%) 17.5 mg (5%) 3.5 mg (1%) 350 mg

Table 1 : Tablets compositions for the 3 API concentrations.

466

80 % 120 %

2.17 2.07 1.92 1.64 2.26

2.14 2.06 1.86 2.01 1.84

M

Disintegration time (s)

Tablet friability KNN

9200-1200

9120-12000

7615-12000 MSC

te

9000-12000

nd

SNV

2 derivative (17pt)

Detrend + Smoothing (15 pt)

3

4

3

0.995 0.415 1.958

0.972 3.87 12.80

0.820 1.67 4.18

Ac ce p

Model Spectral range selected (cm-1) Spectral pretreatment Number of PLS factors R2 val RMSEC RMSEP

Tablet hardness (N) PLS

d

Content uniformity (%)

471

Coefficient of variation (%)

Table 2 : Results of blends content uniformity with the reference method.

468

469 470

Standard deviation (%)

us

A B C

100 %

Mean of sample content uniformity (%) 101.54 100.40 103.32 81.46 123.36

an

Blends

467

120 % % 300 mg (86%) 38.1 mg (11%) 8.4 mg (2%) 3.5 mg (1%) 350 mg

ip t

465

Tables

cr

463 464

N/A

R2 : determination coefficient ; N/A : Not applicable ; all data were mean-centered Table 3 : Conventional criteria of the NIR models.

19 Page 19 of 23

Figure captions

us

cr

ip t

493

Figure 1. Results of real time analysis of the 80% blend. Green points are the reference corresponding to the end of the mixing process when the blend is expected to be homogenous. Blue points are sample spectra of the blend from the beginning to the end of the mixing process.

498 499 500 501

Figure 2. Results of real time analysis of the 100% blend. Green points are the reference corresponding to the end of the mixing process when the blend is expected to be homogenous. Blue points are sample spectra of the blend from the beginning to the end of the mixing process.

Ac ce p

te

d

M

an

494 495 496 497

21 Page 20 of 23

ip t cr

Figure 3. Results of real time analysis of the 120% blend. Green points are the reference corresponding to the end of the mixing process when the blend is expected to be homogenous. Blue points are sample spectra of the blend from the beginning to the end of the mixing process.

us

502 503 504 505

509 510 511 512

Ac ce p

507 508

te

d

M

an

506

Figure 4. Pure API (in green) and tablet (in blue) NIR spectra.

Figure 5. Accuracy profile based on the validation results of the NIR model for the API content determination. The plain line is the relative bias, the dashed lies are the β-expectations tolerance limits (β=95%) and the dotted lines represent the acceptance limits (±10%).

22 Page 21 of 23

ip t cr

Figure 6. Accuracy profile based on the validation results of the NIR model for the tablet hardness determination. The plain line is the relative bias, the dashed lines are the β-expectations tolerance limits (β=95%) and the dotted lines represent the acceptance limits (±25%).

517 518 519

Figure 7. Disintegration time (s) predicted by the NIR model versus the disintegration time measuredby the reference method results for calibration (in red) and validation (in black) sets.

520 521

Figure 8. Results of KNN analysis for tablet friability.

Ac ce p

te

d

M

an

us

513 514 515 516

522 23 Page 22 of 23

Ac

Blenduniformity

ce pt

ed

M an

us

cr

ip t

*Graphical Abstract

Content uniformity

Tablet friability

Disintegration time

Tablet hardness

Real time release approach with:

NIR Spectroscopy

Page 23 of 23

Towards a real time release approach for manufacturing tablets using NIR spectroscopy.

The aim of this study was to use the near-infrared spectroscopy (NIRS) as a process analytical tool to evaluate the conformity of paracetamol tablets ...
2MB Sizes 0 Downloads 3 Views