Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties

Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties

Accepted Manuscript Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties Tinne Monteyne, Jo...

1MB Sizes 4 Downloads 118 Views

Accepted Manuscript Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties Tinne Monteyne, Jochem Vancoillie, Jean-Paul Remon, Chris Vervaet, Thomas De Beer PII: DOI: Reference:

S0939-6411(16)30327-7 http://dx.doi.org/10.1016/j.ejpb.2016.07.021 EJPB 12259

To appear in:

European Journal of Pharmaceutics and Biopharmaceutics

Please cite this article as: T. Monteyne, J. Vancoillie, J-P. Remon, C. Vervaet, T. De Beer, Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties, European Journal of Pharmaceutics and Biopharmaceutics (2016), doi: http://dx.doi.org/10.1016/j.ejpb.2016.07.021

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.

Continuous melt granulation: Influence of process and formulation parameters upon granule and tablet properties Tinne Monteynea,1 , Jochem Vancoilliea , Jean-Paul Remonb , Chris Vervaetb , Thomas De Beera a Laboratory

of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium b Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium

Abstract The pharmaceutical industry has a growing interest in alternative manufacturing models allowing automation and continuous production in order to improve process efficiency and reduce costs. Implementing a switch from batch to continuous processing requires fundamental process understanding and the implementation of quality-by-design (QbD) principles. The aim of this study was to examine the relationship between formulation-parameters (type binder, binder concentration, drug-binder miscibility), process-parameters (screw speed, powder feed rate and granulation temperature), granule properties (size, size distribution, shape, friability, true density, flowability) and tablet properties (tensile strength, friability, dissolution rate) of four different drug-binder formulations using Design of experiments (DOE). Two binders (polyethylene glycol (PEG) and Soluplusr ) with a different solid state, semi-crystalline vs amorphous respectively, were combined with two model-drugs, metoprolol tartrate (MPT) and caffeine anhydrous (CAF), both having a contrasting miscibility with the binders. This research revealed that the granule properties of miscible drug-binder systems depended on the powder feed rate and barrel filling degree of the granulator whereas the granule properties of immiscible systems were mainly influenced by binder concentration. Using an amorphous binder, the tablet tensile strength depended on the granule size. In contrast, granule friability was more important for tablet quality using a brittle binder. However, this was not the case for caffeinecontaining blends, since these phenomena were dominated by the enhanced compression properties of caffeine Form I, which was formed during granulation. Hence, it is important to gain knowledge about formulation behavior during processing since this influences the effect of process parameters onto the granule and tablet properties. Keywords: Continuous hot melt granulation (HMG), granule and tablet properties, DOE, solid state binder, drug binder interaction

Preprint submitted to x

July 19, 2016

1

1. Introduction

2

Granulation is an important process step to improve processability, flowability, compactability and con-

3

tent uniformity of the raw materials to be formulated into a final solid dosage form (e.g., tablets). This

4

particle size enlargement process is often done via wet granulation using a solvent (mostly water) to initiate

5

binding between powder particles, followed by a drying step. However, some pharmaceutical actives are not

6

suitable for wet processing and drying because of stability and degradation problems. In this case, HMG

7

or thermoplastic granulation is a valuable alternative to overcome that problem [1][2]. HMG uses a molten

8

binder instead of a granulation liquid to agglomerate the pharmaceutical powder particles and, hence, no

9

drying step is needed, reducing process time and energy consumption.

10

11

The pharmaceutical industry has traditionally relied on batch processing to manufacture their drug

12

products. However, to improve process efficiency and reduce manufacturing costs, there is a growing inter-

13

est in alternative manufacturing models which allow automation and continuous production. Continuous

14

manufacturing processes are based on the one-in-one-out principle, avoid scale-up issues, ensure faster prod-

15

uct release, reduce production time/costs, reduce variability, increase flexibility and efficiency, and might

16

improve product quality [3][4]. Twin-screw wet granulation has already been examined extensively as con-

17

tinuous granulation technology, allowing fully continuous from-powder-to-tablet-manufacturing [5][6][7][8].

18

However, the research of twin-screw hot melt granulation is still in its infancy.

19

20

Implementing a switch from batch to continuous processing could be perceived as a challenge since this

21

requires fundamental process understanding. The Food and Drug Administration (FDA) published the pro-

22

cess analytical technology (PAT) and QbD concept in the 2004 guidance which were more broadly described

23

in the ICH guidelines Q8, Q9, and Q10 [9][10][11]. The initiative emphasizes the need to understand all

24

critical sources of variability (i.e. critical quality attributes (CQAs), critical material attributes (CMAs) and

25

critical process parameters (CPPs)) affecting a pharmaceutical process to ensure that drug products with

26

predefined quality attributes are consistently obtained. The implementation of QbD principles ultimately

27

aims at designing and developing formulations and processes to build quality into the end product (instead ∗ Corresponding

author Email addresses: [email protected] (Tinne Monteyne ), [email protected] (Jochem Vancoillie), [email protected] (Jean-Paul Remon), [email protected] (Chris Vervaet), [email protected] (Thomas De Beer) 1 Phone number: +32(0)9 264 80 68 2 Fax number: +32(0)9 264 82 36

2

28

of testing quality of the end product). Furthermore, this will enable stable operation, help to support feed-

29

back/feed forward controls, and resulting in fewer compliance problems since a manufacturer anticipates

30

these issues and addresses them more systematically if they occur [12].

31

32

The critical sources of variability are still not fully understood for the continuous melt granulation pro-

33

cess. It is known that the melt granule properties depend on the combination of several process and material

34

parameters and can affect subsequent processing steps, such as milling and tableting. For example, changes

35

in screw speed and configuration (number and location of transport and kneading elements), powder feed

36

rate, temperature, and formulation (type binder, concentration binder, drug-binder interaction) can induce

37

changes in granulation mechanism yielding different granule characteristics which will alter the tablet proper-

38

ties. Ultimately, this can affect the drug dissolution profiles and therefore the bioavailability of the final drug

39

product in a patient [13][14]. This emphasizes the need to define influencing parameters such as CMAs and

40

CPPs on CQAs and can be derived using an experimental design, which after optimization assists in creating

41

a design space for the product [15][16]. However, this has proven to be challenging due to the complex rela-

42

tionships between the granule properties (size, size distribution, shape, solid state, granule friability/strength

43

etc.), and the properties of the final product (tablet tensile strength, tablet friability, dissolution rate) [17][7].

44

45

Literature demonstrated that both the interaction between the initial materials and the type of meltable

46

binder (hydrophilic, hydrophobic, (semi)-crystalline, amorphous) used during the melt granulation process

47

are determining factors for the granulation mechanism [18][19]. Hence, these might have an impact on the

48

process and the final product quality [20]. The aim of this study was to examine the relationship between

49

the formulation parameters (type binder, binder concentration, drug-binder interaction), process-parameters

50

(screw speed, powder feed rate and temperature), granule properties (size, size distribution, shape, friabil-

51

ity, true density, flowability) and tablet properties (tensile strength, friability, dissolution) of four different

52

drug-binder formulations by using DOE. Two binders (PEG4000 and Soluplusr ) with a different solid state

53

(semi-crystalline vs. amorphous) were combined with two model-drugs, MPT and CAF, both having another

54

miscibility with both binders. Four screening DOEs were performed to enhance process understanding and

55

to define the critical material attributes and potential critical process parameters that impact intermediate

56

and final product quality attributes.

57

3

58

2. Materials and methods

59

2.1. Materials

60

PEG and Soluplusr (SLP) were used as hydrophilic meltable binders to develop immediate release for-

61

mulations. PEG4000 (BUFA, Uitgeest, Holland) is a semi-crystalline polymer with a melting temperature

62

(Tm ) of 53 ◦ C. Soluplusr (BASF, Ludwigshafen, Germany) is an amorphous polymer with a glass transi-

63

tion temperature (Tg ) of 70 ◦ C. The binders were used to agglomerate two model drugs, being MPT and

64

caffeine anhydrous. MPT (UTAG, Almere, The Netherlands)(Tm =120 ◦ C) was used as miscible model drug

65

in both binders. MPT has a solubility of 0.53 g per gram PEG4000 whereas it has a solubility of 0.72 g per

66

gram Soluplusr . MPT is a micronized powder having a particle size below 10 µm (X10=1 µm, X50=2.2 µm,

67

X90=7 µm). Caffeine anhydrous (BASF, Ludwigshafen, Germany)(Tm =236 ◦ C), on the other hand, was

68

used as a non-miscible model drug in both binders. CAF is insoluble in PEG4000 whereas it has a solubility

69

of 0.03 g per gram Soluplusr . The solubility parameters of both drugs were obtained from BASF and were

70

based on the Hansen solubility parameter and the Flory-Huggins solubility parameter estimation [21]. Caf-

71

feine has a particle size distribution below 150 µm (X10=1.4 µm, X50=11.1 µm, X90=106.6 µm) and appears

72

in two polymorphs, namely Form I and Form II, the latter being the commercially available polymorph [22].

73

Four different formulations were made: MPT/PEG, MPT/SLP, CAF/PEG, and CAF/SLP. Aerosilr 200

74

(Evonik Degussa Corp., Essen, Germany) was added to the initial powder blend (0.2% (w/w)) to increase

75

the flow properties and hence supporting the feeding into the twin-screw granulator. Before tablet compres-

76

sion, the granules in the size range 150-1400 µm were isolated and both magnesium stearate (0.5% (w/w))

77

and Explotabr (5% (w/w)) were added.

78

79

2.2. Continuous hot melt granulation

80

Melt granulation was performed using a co-rotating intermeshing twin-screw granulator (Prism Eurolab

81

16) (Thermofischer Scientific, Karlsruhe, Germany) with a barrel length of 25 L/D, where L is the axial

82

screw length of the machine and D is the inner bore diameter. The screw design was identical for all

83

experiments containing one kneading zone located in the fifth segment and consisting of 6 kneading discs

84

positioned at a 60 ◦ stagger angle in reversed direction. At the end of the screws, a screw mixing element

85

was placed in order to break up large lumps [23]. The premixed samples were fed into the granulator using

86

a DD Flexwallr 18 gravimetric feeder (Brabender Technologie, Germany), which was set in the gravimetric

87

feeding mode. The barrel was divided into 6 barrel zones. Barrel temperature from segment 2 to 5 was 4

88

varied according to the design. Segment 6, which is located at the end of the barrel, had a lower temperature

89

of 40 ◦ C during all runs in order to cool down the granules and hence avoiding them to stick together when

90

leaving the granulator. Granule samples were collected after melt granulation of each run. The sample

91

collection was started after 15 minutes, which is the time needed to reach a steady state process and a stable

92

torque and barrel wall temperature which were unstable at the beginning of each process due to layering of

93

the screws and the screw chamber walls with material. Sample collection was executed until 150 g sample

94

was collected. Afterwards, samples were stored at room temperature. The granulator was equipped with

95

a data logging system allowing monitoring of the screw torque and actual barrel wall temperature during

96

granulation. The torque (expressed as a percentage, which reflects on the fraction of the maximum torque

97

(12 Nm) that can be generated by the screw motor to rotate the screws) was recorded directly from the

98

twin-screw granulator panel every 30 s. The torque at steady state was averaged to give a single value.

99

2.2.1. Differential scanning calorimetry

100

Differential scanning calorimetry (DSC) was used to measure the melt enthalpy of the pure components,

101

physical mixtures and the corresponding granule samples obtained after melt granulation of the caffeine-

102

containing blends. The data were used to determine the remaining amount of caffeine polymorphic Form II

103

after melt granulation according the equation described by Hubert [24]:

% of caffeine Form II = 104

∆trans Hm(II→I) .100 ∆trans Hm(II→I) of pure Form II

(1)

where ∆trans Hm(II→I) is the enthalpy of transition of the sample.

105

106

A DSC Q2000 calorimeter (TA Instruments, Zellik, Belgium) was used. Samples were hermetically sealed

107

in aluminium pans and a heating rate of 10 ◦ C/min was applied. Analysis was performed in triplicate.

108

2.3. Design of experiments

109

Preliminary experiments were carried out for each formulation to determine the experimental ranges for

110

the DOE factors throughput, screw speed, temperature and binder concentration. The extreme parameter

111

settings where only powder was produced or where an excessive amount of lumps were created after twin-

112

screw hot melt granulation (TS HMG), were eliminated. Four two-level full factorial screening designs (24 )

113

were created (one for each formulation) and executed in order to evaluate the influence of the tested parame-

114

ters on granule and tablet properties. Four factors (powder feed rate, screw speed, binder concentration and 5

115

temperature) were used in each design. Three center points were executed to evaluate the reproducibility.

116

The different factor settings for each design are listed in table 1. The responses were regressed against the

117

factors via multiple linear regression (MLR) (using MODDE 10.0 software by Umetrics, Umea, Sweden). Table 1: Factor settings of the four experimental designs

Settings MPT/PEG MPT/SLP CAF/PEG CAF/SLP

throughput (kg/h) −1 0 +1 0.35 0.625 0.9 0.4 0.6 0.8 0.5 0.8 1.1 0.4 0.7 1

screw −1 100 200 150 200

speed (rpm) 0 +1 225 350 312.5 425 225 300 312.5 425

temperature −1 0 30 44 30 50 55 67.5 50 90

( ◦ C) +1 58 70 80 130

−1 5 5 7.5 5

% binder 0 +1 12.5 20 10 15 15 22.5 12.5 20

118

The influence of the factors is measured on both granule properties (granule friability, true density,

119

compressability index, size (fines, yield, oversized, X50), shape (A50), and span distribution) process torque,

120

and tablet properties (tensile strength, tablet friability, and dissolution rate in 5 minutes).

121

2.3.1. Effect and interactions of factors

122

For each design, the effects were calculated representing the response deviation when a factor is varied

123

from its low level (-1) to its high level (+1) keeping the other factors at their center points. The effects

124

were calculated with their respective 95% confidence interval. Insignificant effects are those where their

125

confidence intervals includes zero. Interaction plots has been calculated to study the predicted change in

126

the response when one factor varies, while a second factor is set at both its low (-1) and high (+1) level,

127

while all other factors being set at their center point.

128

2.4. Granule size distribution and granule shape

129

Size and shape of the final granules were determined (n=3) using an off-line image analysis tool (QicPic,

130

Sympatec, Germany). This high speed imaging tool is based on shadow projection and exists of a VIBRI

131

vibrational conveyor, which is a controlled feeding system, allowing the granules to fall through a shaft where

132

the granules are accelerated and finally pass a laser beam and camera, which records images of shadows of

133

the granules. In this configuration, the system is capable of capturing particles in the range of 10 µm to

134

10.2 mm. Size analysis was based on the equivalent projection of a circle (EQPC). The entire size distri-

135

bution was studied where the size values were classified into 39 classes between 0 and 3400 µm and 1 class

136

>3400 µm. The X10, X50 and X90 values represent the particle size at respectively, 10, 50, and 90% of

137

the cumulative distribution. The X50 values were used as a response in the designs to show the influence

138

of the factors on the granule size. The granule shape was represented by evaluating the aspect ratio which 6

139

measures the elongation of the granule. It is the ratio of the length of the minor and the major diameter of

140

the granule. The granule shape is expressed as A50, representing the aspect ratio at 50% of the cumulative

141

distribution. The width of the size distribution was incorporated as a response in the designs using the span

142

values ((X90-X10)/X50).

143

144

2.5. Granule density

145

The true density (TD) (g/m3 ) of the granules was determined (n=3) using an AccuPyc 1330 helium

146

pycnometer (Micromeritics Instruments Inc, Norcross, United States). A precisely measured amount of

147

sample was placed in the test cell, filling at least 65% of the cell. The pycnometer determined the volume

148

occupied by the sample via pressure changes.

149

2.6. Granule flow properties

150

The compressibility index (CI) describes the powder flow characteristics and can be determined by

151

measuring the bulk and tapped volume of the sample (n=3). The bulk volume (V0 ) of 30 g granules was

152

recorded in a 100 ml measuring cylinder, as well as the volume after 10 (V10 ), 500 (V500 ) and 1250 (V1250 )

153

taps in a tapping machine (J. Englesman, Ludwigshafen, Germany). If the difference between V500 and

154

V1250 was larger than 2 ml, the powder was subjected to another 1250 taps (V2500 ). The compressibility

155

index (%) was calculated from the bulk and tapped density using the following equation:

CI = ((ρi − ρf )/ρi )100 156

157

(2)

where ρi is the bulk density and ρf is the tapped density. 2.7. Friability of granules

158

The granule friability was determined (n=3) using a friabilator (PTF E Pharma Test, Hainburg, Ger-

159

many) at a speed of 25 rpm for 10 min, by subjecting 10 g of granules (Iwt ) together with 200 glass beads

160

(mean diameter of 4 mm) to falling shocks. Prior to determination, the granule fraction <250 µm was re-

161

moved to assure the same starting conditions for all evaluated granules. Afterwards, the glass beads were

162

removed and the weight (g) retained on a 250 µm sieve (Fwt ) was determined. The friability (%) was then

163

calculated as:

F riability = ((Iwt − Fwt )/Iwt )100 7

(3)

164

2.8. Tablet production

165

The granule fraction between 150 and 1400 µm was blended with 0.5% (w/w) magnesium stearate and

166

5% Explotabr in a tumbling mixer (W.A. Bachshofen, Basel, Switzerland). The tablets (115±5 mg) were

167

produced automatically using an eccentric tablet press (Korsch EKO, Berlin, Germany) equipped with a

168

concave punch of 7 mm diameter. The displacement of the upper punch into the die was controlled, resulting

169

in a compression force of 14.7 kN per tablet.

170

2.9. Tablet evaluation

171

The tablet friability was determined (n=3) by subjecting 20 dust free tablets (weight=Iwt ) to falling

172

shocks in a drum rotating at 25 rpm for 4 minutes. The tablets were dedusted and weighed (Fwt ). The per-

173

centage weight loss calculated with equation 3 is expressed as tablet friability and ideally does not exceed

174

1.0%.

175

176

177

The hardness, thickness and diameter of tablets (n=10) were determined (Sotax HT 10, Basel, Switzerland). The tablet tensile strength (TS) (Pa) was calculated using the equation [25]:

T S = 2F/π.dt 178

179

(4)

where F, d and t denote the diametrical crushing force or hardness, the tablet diameter and the tablet thickness, respectively.

180

181

Dissolution tests were performed (n=3) in 900 ml demineralised water using the paddle method (VK

182

7010, Vankel, Cary, NC, USA). The temperature of the dissolution medium was maintained at 37±0.5 ◦ C,

183

while the rotation speed was set at 100 rpm. 5 ml samples were withdrawn at 5, 10, 15, 20, 30, 45, 60, and

184

90 min after starting the dissolution. The drug content was determined at 222 nm for MPT and 273 nm for

185

CAF using an UV-1650PC double beam spectrophotometer (Shimadzu Benelux, Antwerp, Belgium).

186

187

3. Results

188

3.1. Factor ranges

189

The results revealed that the required granulation temperature range was shifted to lower temperatures

190

when the binder and active pharmaceutical ingredient (API) were miscible. This can be related to the 8

191

plasticizing effect of the drug on the binder lowering the temperature at which deformability is initiated.

192

Furthermore, an improved blend deformability during processing enlarged the bonding or contact area of

193

the nuclei when colliding, resulting in plastic deformation which resisted the breakup forces [26][27]. Hence,

194

deformability stimulated granule growth and as a result, the maximum process temperature was lower to

195

avoid overgranulation. On the other hand, when binder and API were immiscible, the maximum granulation

196

temperature exceeded the Tm or Tg of the binder as there was no plasticizing effect of the drug on the binder

197

which resulted in low-deformable granules. In this situation, granule coalescence could only occur if there

198

was a binder layer established at the surface of the particles or granules to bind them [27]. Hence, higher

199

temperatures were required to initiate binder distribution and large lumps were only formed at elevated

200

process temperature.

201

202

3.2. Responses

203

3.2.1. Granule properties

204

The comparison of the granule properties obtained after executing the four designs revealed differences

205

caused by the type of binder, type of API and extent of drug-binder miscibility. The SLP granules were

206

larger compared to the particle size of the PEG granules (table 2). PEG is a semi-crystalline polymer

207

having more elastic properties which can result in more brittle agglomerates compared to SLP, which is an

208

amorphous binder. These brittle agglomerates were not able to withstand the impact of the screw mixing

209

elements at the end of the barrel. The temperature in the last barrel segment was lowered allowing granules

210

already to solidify. Hence, the brittle granules break up more easily resulting into smaller granules. The

211

amorphous binder SLP, on the other hand, produced larger granules, even though when a lower % of binder

212

was used. This implies a larger binding capacity of SLP compared to PEG [28].

213

214

Besides this, the maximum size of the caffeine granules was larger than the size of the granules made

215

with MPT. Several explanations can be found. On the one hand, this might be due to the deviating initial

216

particle size of both APIs. The primary particle size of caffeine reached to 106 µm whereas MPT had an

217

initial size less than 10 microns. Smaller starting material results in smaller particles of the end product

218

as also reported by El Hagrasy and coworkers [29][30]. On the other hand, this might be due to the poly-

219

morph transition from the commercial available caffeine Form II to the metastable Form I, which can occur

220

during twin-screw melt granulation [31]. The metastable Form I has a higher deformability and improved

221

compaction properties favoring the compaction behavior during processing and, hence, stimulating granule 9

Table 2: Absolute values of min, mean and max particle size ( µm), true density (g/m3 ) and granule friability (%) in the four designs

Property Size (X50)

True density

Friability

Value Min. Mean Max. Min. Mean Max. Min. Mean Max.

MPT/PEG 282 675 956 1.213 1.217 1.222 15.6 29.7 43.3

MPT/SLP 363 1137 1700 1.170 1.193 1.214 5.1 13.3 31.6

CAF/PEG/ 478 832 1464 1.399 1.422 1.451 13.1 25.8 43.3

CAF/SLP 299 1092 2128 1.382 1.417 1.458 4.0 34.5 90.7

222

growth. The polymorphic transition to caffeine Form I could also be observed in this research and was more

223

expressed at higher binder concentrations (See below in Figure 11). Furthermore, the conversion was more

224

expressed when Soluplusr was used as a binder. Besides the initial drug particle size and the polymorphic

225

transition, also the granulation mechanism can attribute to the difference in granule size of the caffeine-

226

containing blends in comparison with the MPT-containing blends. During granulation of an immiscible

227

drug-binder blend, the binder behaves as a separate phase which distributes over the drug particles when

228

softened/molten [32]. This distinct binder distribution results in successful coalescence and hence to large

229

granules. Note that process parameters inducing a good binder distribution are necessary to reach successful

230

coalescence. In contrast, interaction (hydrogen bond formation) between drug and binder was discovered

231

during granulation of a miscible drug-binder formulation (MPT/SLP) which impeded proper binder distri-

232

bution making coalescence less evident [20]. In figure 1, a visualization of the granulation mechanism is

233

given based on the observations in previous work.

234

Immersion Drug Binder (ini+al step)

Immiscible drug-binder blend

0 0

0 0 0 0 4 Miscible drug-binder blend

Figure 1: Visualization of granule growth mechanism for a miscible and immiscible binder formulation during twinscrew hot melt granulation.

10

235

The true density measurements (Table 2) revealed that the caffeine-containing granules were more dense

236

(1.4) compared to MPT-containing agglomerates (1.2). Table 4 demonstrates that the granule density was

237

negatively influenced by binder concentration in the two caffeine-containing blends. Therefore, there can be

238

assumed that the higher true density is attributed to the different material properties of caffeine compared

239

to MPT. Furthermore, the conversion of caffeine anhydrous into the more deformable and compactable

240

Form I during granulation could have increased the true density of the granules (Figure 11). On the other

241

hand, it has already been demonstrated that drug-binder miscibility produced more porous granules [33].

242

PEG-containing granules were more fragile which can be related to the brittle properties of the binder. If

243

PEG was used as a binder, the friability was in all cases above 13.1% (CAF/PEG) or 15.6% (MPT/PEG)

244

compared to 4% for CAF/SLP and 5.1% for MPT/SLP. It has to be highlighted that a high temperature

245

or shear was required in order to induce granule growth of the CAF/SLP blend [32]. If not, a high fraction

246

of fines and high friability values were obtained which can be seen in table 2.

247

248

3.2.2. Tablet properties

249

The tablet properties revealed that tablets made with Soluplusr have the highest maximum tablet tensile

250

strength, with values of 3.73 MPa for CAF/SLP and 3.33 MPa for MPT/SLP. In comparison, CAF/PEG

251

and MPT/PEG have a maximum tablet tensile strength of 2.44 and 2.66 MPa, respectively. The more

252

ductile behavior of Soluplusr allowed plastic deformation of the granules during tableting, yielding stronger

253

tablets [34]. Besides tensile strength, also dissolution rate is different for both binders. When the API

254

is granulated at the same granulation conditions with both PEG and Soluplusr , the dissolution rate will

255

be 10% faster after granulation with PEG (for both MPT and CAF). Furthermore, when PEG is used to

256

agglomerate caffeine, the entire drug content is dissolved in 30 minutes, whereas only 70% caffeine is released

257

from Soluplusr mixtures. Earlier research on melt granulation already showed PEG to be a suitable binder

258

to enhance the dissolution rate of poorly water-soluble drugs [35][36]. The higher hydrophilicity of PEG

259

might be a possible explanation.

260

261

262

A list of significant effects of the factors and their interactions is given in tables 4 (granule properties) and 5 (tablet properties).

11

Table 3: Absolute values of min, mean and max tensile strength (MPa) and amount API (%) released in 5 minutes for the four designs

Property Tensile strength

API (%) released in 5 min

263

3.3. Influence of powder feed rate

264

3.3.1. Granule properties

Value Minimum Mean Maximum Minimum Mean Maximum

MPT/PEG 1.18 1.93 2.66 61.4 72.4 84.2

MPT/SLP 1.41 2.59 3.33 33.3 63.2 78.9

CAF/PEG 1.19 2.15 2.44 17.3 27.7 52.7

CAF/SLP 1.11 2.02 3.73 8.3 18.3 28.5

265

The influence of powder feed rate on the granule properties depended on the API. In general, throughput

266

had a large influence on the MPT-containing granules, whereas it had a minor effect on the caffeine-containing

267

granules (Table 4). Powder feed rate had even no influence on the granule properties of the CAF/SLP blend.

268

This is in accordance with the miscibility parameters of the APIs with the polymers, except for CAF/PEG.

269

It has to be highlighted that every run in the design of CAF/PEG was performed using a barrel temperature

270

exceeding the melting temperature of PEG and hence it can be stated that PEG was molten throughout

271

the whole design. Therefore, it is obvious that the enhanced compressive forces obtained at high powder

272

feed rate affected the granulation process of CAF/PEG, regardless the miscibility of both materials. The

273

importance of powder feed rate for the granulation properties of miscible systems could be found in the

274

specific granulation mechanism. In miscible drug-binder formulations, granule growth occurs when nuclei

275

deform after they collided. The nuclei are rather ‘kneaded’ instead of sticking together as was seen for

276

immiscible binder blends (Figure 1).

277

278

Performing granulation with a higher throughput resulted in larger and stronger granules with improved

279

flow properties. These effects were negligible for the CAF/SLP blend and less pronounced for CAF/PEG

280

blend (Table 4). These observations might be attributed to the higher screw fill in the barrel appearing

281

at increasing powder feed rate and constant screw speed. The degree of channel fill refers to the volume

282

occupied by the powder blend with respect to the total available volume in the conveying zones of the screw.

283

A higher powder feed rate induces superior screw filling, generating a high throughput force which conveys

284

the powder more quickly through the barrel [37][38]. As a result, the residence time of the material inside

285

the barrel is shortened, but the material packing and compressive forces in the material between the screws

286

is enhanced. The latter induce more interactions between the powder particles or the yet developed gran12

13

T*%bi

Scr*T

Scr*%bi

Thr*T

Thr*%bi

Thr*Scr

Temperature

Binder %

Screw speed

Throughput

Factor

Friability

-5.09 (±3.84) -8.08 (±3.47) -7.6 (±4.39) 24.15 (±18.5) -15.87 (±3.84) -5.07 (±3.47) -15.90(±4.39) -39.46 (±18.5) 4.3 (±3.47) -28.15 (18.5) -9.28 (±3.47) 5.13 (±4.39) 3.47 (±3.47) 5.07 (±3.84) -5.4 (±3.47) -6.69 (±4.39) 6.68 (±3.47) 6.79 (±3.84) 7.90 (±3.47) -

Design

MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP

-0.0175 (±0.01) -0.042 (±0.0043) -0.056 (±0.006) -

Density -3.61 (±2.85) -6.7 (±5.38) 8.70 (±6.40) -3.68 (±2.85) -5.91 (±5.38) -5.35 (±2.92) 2.95 (±2.92) -7.30 (±6.40)

Comp. index -5.59 (±2.2) -9.75 (±4.34) 9.76 (±4.34) 17.25 (±12.5) -10.17 (±2.2) -8.99 (±6.33) -16.51 (±12.50) -2.89 (±2.2) -9.97 (±4.34) 4.11 (±2.2) -

Fines 5.63 (±4.05) -15.12 (±6.85) -7.56 (±5.11) 13.75 (±5.11) 17.51 (±4.05) 4.45 (±4.05) -15.64 (±6.85) -

24.86 (±10.23) 11.6 (±10.7) -13.95 (±10.23) -16.70 (±10.7) 12.09 (±10.7) 32.07 (±17.40) 10.94 (±10.35) 25.61 (±10.23) -

Granule properties Yield Oversized 276.97 (±146.43) 578.92 (±228.77) 270.1 (±226.96) -256.96 (±228.77) -257.71 (±226.96) 348.299 (±226.96) 627.57 (±575) 630.117 (±575) 577.58 (±228.77) -

X50

-0.0422 (±0.022) -0.017 (±0.0127) 0.026 (±0.022) 0.0275 (±0.018) 0.045 (±0.043) -0.0175 (±0.0127) -0.054 (±0.022) 0.015 (±0.0127) -

A50

Table 4: List of significant effects of the factors and their interactions on granule properties of the four designs: granule friability (%), true density (g/m3 ), compressability index (CI)(%), fines (%), yield (%), oversized (%), X50 (µm), A50 (-) and SPAN (-). SPAN -0.44 (±0.39) -0.28 (±0.19) 0.20 (±0.19) -0.51 (±0.39) -2.54 (±0.92) -0.34 (±0.19) -1.14 (±0.92) -

Table 5: List of significant effects of the factors and their interactions on process and tablet properties of the four designs: torque (%), tensile strength (MPa), tablet friability (%) and % drug released after 5 minutes.

Factor Throughput

Screw speed

Binder %

Temperature

Thr*Scr

Thr*%bi

Thr*T

Scr*%bi

Scr*T

T*%bi

Design

Process property Torque

Tensile strength

MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP MPT-PEG MPT-SLP CAF-PEG CAF-SLP

9.06 (±2.05) 4.375 (±2.41) -14.19 (±2.05) -10.125 (±2.41) -22.29 (±7.73) 10.29 (±7.73) -3.062 (±2.05) -4.06 (±2.05) 2.88 (±2.41) -2.06 (±2.05) -

0.58 (±0.36) -0.483 (±0.24) -0.56 (±0.36) -0.51 (±0.24) 0.4 (±0.20) 0.93 (±0.74) -0.36 (±0.24) 0.5 (±0.36) -0.27 (±0.24) 0.39 (±0.36) -0.36 (±0.24) 0.536 (±0.24) 0.83 (±0.74) 0.78 (±0.74)

14

Tablet properties Friability -3.24 (±2.61) -1.0 (±0.84) -2.16 (±1.4) -4.79 (±2.61) -1.41 (±1.4) -0.89 (±0.84) 3.36 (±2.61) -

Release -11.86 (±8.29) -9.40 (±3.96) -11.7 (±3.73) -20.37 (±8.29) 5.44 (±3.73) 9.6 (±8.29) 10.75 (±8.29) -

287

ules and the ungranulated powder, causing the powder to efficiently attach on the surface of the granules

288

or around each other which stimulates granule growth. The higher the powder feed rate, the better the

289

interlocking of the primary particles onto the structure of the granules, the stronger the granules become.

290

As a consequence, lower friability rates were obtained when a high powder feed rate was used (Table 4).

291

Furthermore, it is evidenced that the high material packing facilitated interaction between drug and binder

292

explaining the higher influence of throughput on the granule properties for blends having the possibility of

293

interacting (MPT/PEG and MPT/SLP) [39]. Additionally, the superior screw filling reduced the amount of

294

fines since binding of ungranulated powder particles took place at enhanced compression forces [38]. These

295

outcomes were similar to these obtained in continuous wet granulation [40][41].

296

297

The flow properties of the granules obtained from the miscible blends depended on the amount of fines

298

and the span distribution and therefore they are influenced by powder feed rate (Figure 2). The granules

299

have an improved flowability when the amount of fines is sparse, or/and when the span distribution is narrow

300

which is achieved at elevated powder feed rate. However, the throughput can not be too high in order to

301

limit the amount of oversized granules[42].

302

4

20

3

10

1.5

0

1

-10

0

Y=1.3088x-8.066 R2= 0.55349

10

15

20

25

20

2

Compressability index (%)

10 Y=0.600x-10.02 R2= 0.70549

10

15

20

25

SPAN Fines Fines (%)

2.0

1.0

30

SPAN Y=0.0365x+1.3033 Fines R2= 0.65576 Fines (%)

SPAN (-)

2.5

30 Y=5.79x+0.2055 R2= 0.3934

SPAN (-)

3.0

0

Compressability index (%)

Figure 2: Correlation plots for MPT/SLP (left) and MPT/PEG (right) between compressability index (%) and span distribution (◦) and between compressability index (%) and fines (%)(N).

303

3.3.2. Tablet properties

304

The powder feed rate had almost no influence on the tablet properties of the caffeine-containing gran-

305

ules, which is in accordance with its effect on the granule properties (Table 5). MPT/SLP tablets had a

306

sufficient strength with a friability lower than 1% in all cases. For MPT/PEG containing tablets, on the

307

other hand, capping was observed at parameter settings where adhesion was impeded and is demonstrated

308

in figure 3. When the binder concentration or granulation temperature were low, an increase in powder feed 15

4

Temp high Temp low

3

Tablet friability (%)

Tablet friability (%)

4

2 1 0 0.3

0.6

Binder % high Binder % low

3 2 1 0 0.3

0.9

Powder feed rate (kg/h)

0.6

0.9

Powder feed rate (kg/h)

Figure 3: Interaction plot for tablet friability between (left) powder feed rate and granulation temperature and (right) powder feed rate and binder concentration of the MPT/PEG blend.

309

rate affected tablet friability. Increasing the powder feed rate shortened the material residence time inside

310

the barrel which impeded the binder to melt and, hence, to distribute/bind drug particles. Compressing

311

these granules into tablets led to capping of the tablets [43]. If the binder concentration and/or process

312

temperature was higher, the binding capacity was better making the material residence time inside the

313

granulator or the material throughput less important since under these conditions, tablet friability values

314

remained constant below 0.5% and a good tablet strength was obtained.

315

316

3.4. Influence of screw speed

317

3.4.1. Granule properties

318

Screw speed was influencing the granule size and shape in all four designs. Additionally, there was an

319

interaction between the effects of screw speed and powder feed rate. This interaction is attributed to the

320

extent of screw fill which is correlated to torque. An accelerated screw speed diminished the barrel filling

321

degree and resulted in lower torque values. This interaction was observed for the miscible formulations and is

322

linked to the specific granulation mechanism for miscible formulations as described above (Table 4)(Figure

323

1). Granule growth occurred by kneading nuclei together and therefore, a high barrel filling degree was

324

required. In any case, a correlation between the torque level and amount of fines was observed where higher

325

torque values generated granules containing less fines (Figure 4). The superior channel filling at low screw

326

speed caused adherence of the not-agglomerated powder to the surface of the already formed granules and

327

to each other [38]. As a result, the amount of fines was reduced.

328

329

Furthermore, screw speed affected the granule size distribution of the miscible drug-binder blends, but

330

the effect depended on the powder feed rate. When the powder feed rate was low (−1), a slower screw speed 16

25

Scr.sp. high (CAF/SLP)

Scr.sp. high (CAF/PEG) 20

30

Fines (%)

Torque (%)

40

20 10

Scr.sp. low (CAF/SLP) 15 Scr.sp. low (CAF/PEG)

high (CAF/SLP) high (CAF/PEG) low (CAF/SLP) low (CAF/PEG)

Scr.sp. Scr.sp. Scr.sp. Scr.sp.

high (MPT/SLP) high (MPT/PEG) low (MPT/SLP) low (MPT/PEG)

10 5 0

0 -1

0

-1

+1

0

+1

Powder feed rate (kg/h)

Powder feed rate (kg/h) 40

25

Scr.sp. high (MPT/SLP)

Scr.sp. high (MPT/PEG) 20

30

Fines (%)

Torque (%)

Scr.sp. Scr.sp. Scr.sp. Scr.sp.

20 10

Scr.sp. low (MPT/SLP) 15 Scr.sp. low (MPT/PEG) 10 5

0

0 -1

0

+1

-1

0

Powder feed rate (kg/h)

+1

Powder feed rate (kg/h)

Figure 4: Interaction plot for (left) torque (%) and (right) fines (%) between powder feed rate (kg/h) and screw speed (rpm) of (up) CAF/SLP and CAF/PEG and (low) MPT/SLP and MPT/PEG blends. Soluplusr is shown by the dotted and PEG by the solid lines. (•) and (◦) represent high screw speed, whereas () and () symbol the low screw speed. CAF/SLP

CAF/PEG 50

30

Thr (-1) scr (-1) 40 (+1) scr (-1) Thr Thr (-1) scr (+1) 30 Thr (+1) scr (+1)

(-1) scr (-1) (+1) scr (-1) (-1) scr (+1) (+1) scr (+1)

Thr Thr Thr Thr

(-1) scr (-1) (+1) scr (-1) (-1) scr (+1) (+1) scr (+1)

%

%

20

Thr Thr Thr Thr

20 10

10 0 100

1000

0 100

10000

1000

10000

Granule size (µm)

Granule size (µm)

MPT/SLP

MPT/PEG 40

25 20

%

%

15

Thr Thr 30 Thr Thr 20

(-1) scr (-1) (+1) scr (-1) (-1) scr (+1) (+1) scr (+1)

10

10

5 0 100

1000

10000

0 100

1000

10000

Granule size (µm)

Granule size (µm)

Figure 5: Granule size distribution of runs with varying combinations of throughput and screw speed.

331

produced larger granules with a broader granule size distribution. This is demonstrated in figure 5. At

332

low screw speed, the channel filling degree of the screws (compared to high screw speed) and the residence 17

333

time of the material inside the barrel were enlarged. The additional time of the material inside the granu-

334

lator stimulated granule growth. When the powder feed rate was high (+1), accelerating the screw speed

335

produced more oversized granules, but the amount of fines remained equal. Hence, a broader distribution

336

was obtained. This is linked to the shortened material residence time of the material inside the barrel at

337

high screw speed and high powder feed rate. As a consequence, the screw channels were starved of powder

338

which limited powder compaction [38]. In order to reach an appropriate size distribution, both screw fill

339

degree and residence time inside the barrel must be controlled as they are both playing a key role. The

340

longer the residence time, the higher the collision frequency of the granules with the barrel wall will be.

341

Therefore, a prolonged residence time is required for granules to reduce the oversized fraction. On the other

342

hand, a high screw filling degree is preferred in order to bind the ungranulated powder on the surface of the

343

granular material reducing the fines. This explains the appearance of a fine granular material accompanied

344

by oversized granules when operating at fast screw speed and high powder feed rate.

345

CAF/SLP 50

T (-1) Bi (-1) Scr (-1) T (-1) Bi (-1) Scr (+1) T (+1) Bi (+1) Scr (-1) T (+1) Bi (+1) Scr (+1)

40

%

30 20 10 0 100

1000

10000

Granule size (µm)

Figure 6: Granule size distribution of the SLP/CAF blend when deviating screw speed from −1 to +1 at different levels of binder concentration and granulation temperature.

346

The behavior of the granules obtained from granulation of CAF-containing blends was deviating in

347

specific cases. Performing granulation of the CAF/PEG blend using a higher screw speed was always pro-

348

ducing a higher fraction of smaller sized granules, independent of the powder feed rate (Figure 5). For

349

the CAF/SLP blend, the behavior was deviating when a low binder content and low barrel temperature

350

or high binder concentration and high barrel temperature were used (Figure 6). The influence of powder

351

feed rate was negligible in these cases. Accelerating the screws during granulation of caffeine using a low

352

Soluplusr concentration at a low barrel temperature, produced both small and oversized granules. The

353

reason for this might be the shortened time to soften the limited amount of binder, thus hindering granule 18

354

growth. The few granules were formed via immersion of the solid particles on the surface of the softening

355

binder. Furthermore, a high granulation temperature and/or high binder concentration was required for this

356

blend in order to induce agglomeration [32]. On the other hand, speeding up the screws during granulation

357

of caffeine using a high Soluplusr content at elevated barrel temperature generated an excessive amount

358

of oversized granules. The higher screw tip speed induced mechanical shear which enhanced softening of

359

Soluplusr . Besides improved binder distribution also transition of the stable caffeine polymorphic form

360

to Form I occurred, facilitating granule growth [31](Figure 11). Furthermore, it can be assumed that the

361

granule size distribution depends on the type of binder, type of API and whether they interact or not. This

362

is in contrast with the observations from Dalziel and coworkers who suggested that granule particle size

363

is independent of the binder or drug-polymer ratio and that granule size is a function of the processing

364

techniques [44].

365

366

The influence of screw speed on the granule shape was related to the drug-binder miscibility and on

367

the type of binder (Table 4). When the miscibility between binder and API was sparse (caffeine-containing

368

blends), the produced granules became more needle-shaped when operating at slower screw speed. Due to

369

the prolonged residence time of the material inside the barrel, the binder had more time to distribute over

370

the solid particles. Hence, the shear in the mixing zone elongated the granules rather than fragmenting them.

371

This effect was limited for the CAF/PEG blend due to the less viscous and more brittle properties of the

372

PEG binder. When binder and API had a certain miscibility, a slower screw speed generated more spherical

373

granules. Due to the elongated material residence time inside the barrel, more binder-API interaction took

374

place, hindering the binder to distribute restricting elongation [20]. For the MPT/SLP blend, this could

375

only be observed when operating at high powder feed rate as shown in the interaction plot in figure 7. At

376

high screw speed and low powder feed rate, there was insufficient compaction due to the inadequate filling

377

degree of the barrel resulting in more fines and, thus more spherical granules. At superior barrel filling

378

(at higher powder feed rate) more compaction occurred and the influence of screw speed on granule size

379

was more pronounced. When sufficient compaction occurred for the MPT/SLP blend, a lower screw speed

380

generated more spherical granules as was seen for the for the MPT/PEG blend.

381

382

3.4.2. Tablet properties

383

The influence of screw speed on tablet tensile strength was only significant for the miscible blends where

384

accelerating the screws resulted in tablets with a lowered tensile strength (Table 5). However, the granule 19

25

0.75

0.70

15 10

Low screw speed (Fines) High screw speed (Fines) Low screw speed (A50) High screw speed (A50)

0.65

5 0

A50 (-)

Fines (%)

20

0.4

0.5

0.6

0.7

0.8

Powder feed rate (kg/h)

Figure 7: Interaction plot for (dotted lines) fines (%) and (solid lines) A50 (-) between powder feed rate (kg/h) and screw speed (rpm) of the MPT/SLP blend. The squares represent low screw speed, whereas the circles symbol the high screw speed.

385

properties determining the tensile strength (granule size vs granule friability) were different for brittle and

386

amorphous binders. Using an amorphous binder, the granule size is important regarding tablet quality.

387

Larger granules produced tablets with an enhanced tensile strength. For brittle binders, not size but gran-

388

ule friability should be taken into account to obtain tablets having a sufficient tensile strength. Brittle

389

granules can easily break up during compression, enlarging the available bonding surface stimulating tablet

390

tensile strength. Screw speed is only affecting the tablet tensile strength of the MPT-containing blends. In

391

contrast, caffeine-containing blends yielded stronger tablets when using a higher binder content. This will

392

be explained later.

393

394

Tablets obtained after compression of MPT/SLP granules showed a tensile strength which positively

395

correlated with granule size (X50). The R2 counted 0.7 for the MPT/SLP blend in contrast to only 0.07

396

for the MPT/PEG blend. The relationship between granule size and tensile strength was already observed

397

by Sun and coworkers. However, they explained a reduced tabletability as due to granule size enlargement

398

of plastic material [45]. In this research the opposite relation was observed, since larger granules (X50)

399

resulted in tablets with improved tensile strength. In the design of the MPT/SLP blend, larger granules

400

were obtained at parameters causing a high barrel filling. This stimulated binder and drug to interact

401

causing plasticization of the binder. Hence, one may assume that the larger particles were more deformable

402

compared to smaller granules and, hence, causing tablets with enlarged tensile strength. During compaction

403

of deformable granules, more intermolecular bonding sites are formed over an equal contact area compared

404

to less deformable granules [46]. On the other hand, accelerating the screw speed reduced the barrel filling

405

degree, and thus lowered the binder-drug interactions. As a result, the granules were less deformable during

406

compression generating tablets with a weaker tensile strength. (Table 5). 20

407

408

When granules were made using PEG, the tablet tensile strength was not correlated with the granule

409

size. PEG is a semi-crystalline polymer and is very brittle, in contrast to Soluplusr which is amorphous and

410

therefore more deformable. Since the PEG-containing granules broke-up during compression, the granule

411

friability was important for the tablet tensile strength and not the original granule size. During tablet com-

412

pression, the weak granules were fragmented in a large amount of smaller particles, enlarging the available

413

binding area which improved the tablet tensile strength. The R2 between the granule friability and tablet

414

tensile strength counted 0.62 for the MPT/PEG blend. Due to the negligible influence of screw speed on

415

the granule properties of the CAF/PEG blend, screw speed is not influencing its tablet properties (Table

416

5).

417

418

For the MPT/PEG blend, the influence of screw speed can be found in the interaction screw speed-binder

419

concentration. When granulation is executed using a low binder content, a faster screw speed favors granule

420

strength and, hence, tablets with a reduced tensile strength are generated. This is demonstrated in figure 8.

421

When granulation is performed using a high binder concentration, stronger granules are obtained (relative to

422

the granules having a lower binder content) which produced tablets with a lower tensile strength. At higher

423

PEG concentrations, the granule friability was less influenced by screw speed and more by powder feed rate.

424

The relationship between granule friability and tablet tensile strength was also studied by Wu and coworkers

425

and confirmed these findings [47]. These observations allow to reduce the effects of granule size variations

426

on the tablet properties by incorporating an appropriate amount of brittle binder to the formulation.

427

2.4 2.2

30 2.0 1.8

20 100

200

Tensile strength (MPa)

Friability granules (%)

2.6 40

Low binder conc (friability) High binder conc (friability) Low binder conc (TS) High binder conc (TS)

1.6

300

Screw speed (rpm)

Figure 8: Interaction plot for (dotted line) granule friability (%) and (solid line) tablet tensile strength (MPa) between screw speed (rpm) and binder concentration (%) of the MPT/PEG blend. The circle represents low binder concentration, whereas the triangle symbols the high binder concentration.

428

Additionally, screw speed affected the dissolution rate for the granules manufactured using Soluplusr . 21

429

Speeding up the screws, lowered the % drug released after 5 minutes (11.86% for MPT/SLP and 9.40%

430

for CAF/SLP) (Table 5). This can be linked to higher fraction of fines for these blends when operating at

431

high screw speed and low powder feed rate. The fines and oversized granules were excluded before tablet

432

production. If the fines contain mainly API, this can reduce the diffusion gradient and, hence, the % drug

433

released. This was the case, since the maximum drug release never exceeded 90% and 58%, for respectively

434

MPT/SLP and CAF/SLP, at these parameters (Figure 9). The increase in % fines could be attributed to

435

the limited barrel filling degree when operating at high screw speed and low powder feed rate, as afore-

436

mentioned. Furthermore, since Soluplusr needs to soften in order to allow binding, the shorter material

437

residence time inside the barrel (due to the higher screw speed) will have a superior impact on the Soluplusr

438

blends compared to the PEG blends.

439

CAF/SLP

20

80

15

70

10

60

5

% CAF released

90

100

Thr Thr 80 Thr Thr 60 Thr Thr 40

30

low (release 5min) high (release 5min) low (max release) high (max release) low (fines) high (fines)

20

10

% fines

25

% fines

% MPT released

MPT/SLP 100

Thr Thr Thr Thr Thr Thr

low (release 5min) high (release 5min) low (max release) high (max release) low (fines) high (fines)

20 50

200

300

400

0

200

300

400

0

Screw speed (rpm)

Screw speed (rpm)

Figure 9: Interaction plot for drug release after 5min, max drug release (90min) and fraction fines between screw speed (rpm) and powder feed rate (kg/h) of the (left) MPT/SLP and (right) CAF/SLP blend. The dotted lines represent high throughput, whereas the solid lines symbol low throughput.

440

3.5. Influence of temperature

441

3.5.1. Influence on granule properties

442

Temperature had almost no influence on the granule properties of PEG-containing blends. This can be

443

explained by the fact that PEG is a semi-crystalline polymer having a melting point. When the melting

444

point of a crystalline material is reached, the solid state transfers abrupt from the solid to the molten state.

445

The PEG binder was in the molten state in each run since the actual temperature was close to the melting

446

point throughout both designs. Hence, elevating barrel temperature barely affected the binder viscosity and

447

therefore the influence on the granule properties was negligible.

448

449

Soluplusr on the other hand is an amorphous binder, which softens gradually in function of tempera-

450

ture. A large influence of temperature was seen for the CAF/SLP blend. When granulation of this blend 22

451

was performed at elevated barrel temperature, stronger and larger granules with a smaller span distribution

452

were achieved. In contrast, almost no influence of temperature on the granule properties of the MPT/SLP

453

blend could be observed. This can be attributed to the fact that intermolecular interactions did occur be-

454

tween Soluplusr and MPT impeding binder and API to move as two separate phases. As a result, binder

455

distribution became difficult, and more friable granules were generated [20]. When drug-binder interactions

456

were absent (CAF/SLP), binder distribution was dominated by the viscosity of the binder and was not re-

457

stricted by the interactions. Elevating the granulation temperature, lowered the binder viscosity facilitating

458

distribution. In contrast, when intermolecular interactions did occur (MPT/SLP), binder and API could

459

not move independently and were moving rather as one mass, making viscosity of the binder less important

460

for the granule properties [20]. Furthermore, due to the hydrogen bond formation between MPT and SLP,

461

the maximum useable barrel temperature was 70 ◦ C (plasticizing effect), which is lower than the maximum

462

temperature of 130 ◦ C used for the CAF and SLP blend.

463

464

An improved binder distribution induces better overall binding for all particles favoring granule strength

465

and granule growth. A temperature increase from 50 to 130 ◦ C during granulation of the CAF/SLP blend

466

reduced granule friability by 28%. The reason for the better granule strength due to higher granulation

467

temperature can be twofold. On the one hand, the binder distribution is facilitated at higher temperatures

468

(due to lower viscosity) improving coalescence. On the other hand, caffeine anhydrous can turn into the

469

metastable polymorph (Form I) at high granulation temperature. Caffeine Form I is more deformable in

470

comparison with Form II and improved the granulation efficiency and compaction properties [48][31]. There-

471

fore, the polymorph transition into Form I can favor granule strength. This could be observed during DSC

472

analysis which is shown in figure 11. In contrast, temperature stimulated hydrogen bond formation between

473

MPT and SLP, restricting binder distribution and more fragile granules were obtained (Table 4).

474

475

3.5.2. Influence on tablet properties

476

Temperature was barely influencing the measured granule properties of the MPT/SLP blend, but did

477

affect the tablet properties of this blends. Increasing the granulation temperature facilitated the API-binder

478

interactions which improved the deformability of the granules. Earlier research revealed that interaction

479

between MPT and SLP plasticized the binder (lower Tg ) [43]. Furthermore, at low granulation tempera-

480

ture, granule growth occurred through binding several smaller nuclei together. At elevated temperature, the

481

binder molecules bound together forming larger nuclei which have a higher deformability [20]. During tablet 23

482

compression, the better deformability of the granules initiated a higher bonding capacity per unit area of

483

contact and, hence, the tablet tensile strength improved (Table 5).

484

485

Additionally, the enhanced miscibility occurring at higher barrel temperature stimulated the dissolution

486

rate of MPT into the dissolution medium (Table 5). Possible reasons are the enhanced wettability of the

487

drug when interaction occurred with the binder or the improved solubility of drug when it is in the amor-

488

phous state due to interaction [33]. Since temperature had a limited effect on the granule properties of the

489

MPT/PEG blend, the influence of temperature on dissolution rate was also less pronounced for this blend.

490

It has already been reported that hydrogen bonds were formed between MPT and Soluplusr . These were

491

rather limited at low barrel temperature, but highly expressed at elevated barrel temperature [20]. This

492

explains the pronounced effect of temperature (from Temperature (T ) setting −1 to +1) on the release rate

493

of MPT from the Soluplusr binder (9.6%) in comparison with the release from the PEG binder (5.4%).

494

However, the absolute values of the dissolution rate revealed that the release of MPT from the PEG binder

495

was faster in comparison with the Soluplusr binder which was due to the higher hydrophilicity of PEG (see

496

table 3).

497

498

3.6. Influence of Binder concentration

499

3.6.1. Influence on granule properties

500

In general, more binder generated larger and stronger granules (Table 4). A higher binder concentration

501

strengthens bonds between the drug particles as there is more binder available for bonding. Binder con-

502

centration did not affect the granule size of the MPT/SLP blend. The narrow range of the factor binder

503

concentration in the design of MPT/SLP can be a possible explanation. When granulation was performed

504

using PEG as a binder, increasing the binder concentration formed less fines. In contrast, for the Soluplusr

505

binder, the same effect could be obtained by reducing screw speed. The binding capacity of PEG appears

506

to be lower, and therefore, more binder was needed to bind the unagglomerated particles.

507

508

Melt granulation of the caffeine-containing blends using more binder produced more porous granules,

509

regardless which binder was used (Table 4). The reason for this might not be the binder itself but the lower

510

caffeine concentration (due to higher binder concentration). As aforementioned, the crystalline structure

511

of caffeine anhydrous can modify from the commercial Form II into the metastable and more deformable

512

and more compactable Form I due to the shear and heat during continuous twin-screw melt granulation. 24

513

The higher compactability of Form I can cause a higher density (lower porosity) of the granules when more

514

caffeine is used. As a result of improved compactability, also the fraction oversized granules became larger.

515

516

Executing melt granulation using a high binder concentration produced stronger granules (Table 4).

517

This was also observed by Gokhale et al. during high-shear granulation. Since more binder was used, more

518

bridges were formed between particles, resulting in stronger granules. Although the effect was common,

519

the size of the effect differed for each design. An interaction between temperature and binder concentra-

520

tion could be observed for the blends where interaction occurred between binder and API (MPT/SLP and

521

MPT/PEG)(Table 4). In both designs, an elevated temperature at low binder content yielded stronger

522

granules, due to a lower viscosity and subsequent improved binder distribution and bonding. When using

523

more binder, higher temperatures will yield weaker granules. Binder distribution is impeded due to hydrogen

524

bond formation between binder and API causing more fragile granules (Figure 10). However, these effects

525

were rather low.

526

Friability granules (%)

40

Binder Binder Binder Binder

30 20

% % % %

low (MPT/SLP) high (MPT/SLP) low (MPT/PEG) high (MPT/PEG)

10 0

-1

0

1

Granulation T (°C)

Figure 10: Interaction plot for granule friability (%) between binder concentration (%) and granulation temperature ( ◦ C) of blends MPT/SLP (solid lines) and MPT/PEG (dotted lines).

527

3.6.2. Influence on tablet properties

528

Regardless of the binder used during granulation of MPT, including more binder resulted in tablets with

529

a lower dissolution rate. A higher binder content allowed more binding between particles, on the one hand,

530

but caused a reduction in total amount of drug. Hence, a slower release of the drug molecules was achieved

531

(Table 5). This effect was higher for the fast dissolving MPT and is not significant for the poorly soluble

532

caffeine. Furthermore, the higher binder concentration increased the binding capacity and, hence, tablets

533

with a lower friability were obtained.

534

25

2.0 A

142.56°C 14.70J/g 162.76°C B 139.21°C 14.03J/g

Heat Flow (W/g)

147.86°C

C

143.03°C 11.96J/g 162.58°C D

138.58°C 8.924J/g

144.04°C E

0.2 100 Exo Up

150

200

Temperature (°C)

Universal V4.5A TA Instruments

Figure 11: Heat flow curve of (A) pure caffeine and (B) granules of blends CAF/PEG and (C) CAF/SLP prepared at low binder concentration and (D) granules of blends CAF/PEG and (E) CAF/SLP at high binder concentrations showing the transition of the remaining caffeine Form II into caffeine Form I.

535

Including more binder during continuous melt granulation lowered the tensile strength of the MPT/PEG

536

tablets. This is caused by the enhanced granule strength due to the superior binding derived from the higher

537

binder concentration. Stronger granules underwent less brittle fracture during tablet compression and, hence,

538

a weaker tablet strength was obtained. However, the opposite effect on tensile strength could be observed

539

for the caffeine-containing granules due to the higher deformability after polymorphic transition to the more

540

deformable caffeine Form I. From DSC analysis, it could be observed that more caffeine polymorph Form

541

I was formed when more binder was used during melt granulation (Figure 11). For the CAF/SLP blend,

542

no caffeine Form II could be observed after granulation at higher Soluplusr concentrations, whereas still

543

60% caffeine Form II was detected after granulation with high PEG concentrations. Hence, the effect of

544

binder concentration on tablet tensile strength was more expressed for the CAF/SLP blend. Furthermore,

545

since Soluplusr is ductile, increasing the concentration also generates deformable granules which maximizes

546

interparticle bonding per unit of contact area [49].

547

548

4. General conclusion

549

This research revealed that the type of binder, type of API and interactions between both affect the

550

granulation mechanism of the blend during the continuous melt granulation process and, hence, determine 26

551

the relationship between the influencing parameters and the CQAs (See summarizing table 7). Drug-binder

552

miscibility induces plasticizing of the binder which limited the maximum possible granulation temperature.

553

For these blends, granule growth was rather attained by kneading granules together and, thus the extent of

554

barrel filling was highly influencing the granule and tablet properties. Granule growth of immiscible drug-

555

binder blends was acquired after distribution of the binder allowing nuclei to stick together. Since the binder

556

did behave as a separate phase, the granulation temperature could exceed the Tg /Tm of the binder. For these

557

blends, binder concentration was the most influencing factor. Furthermore, these blends contained caffeine

558

anhydrous as a drug, which is able to convert from the commercial Form II into the more deformable Form

559

I, which facilitated granule growth. This conversion was favored at elevated granulation temperature and/or

560

higher binder concentration and was more expressed when Soluplusr was used as a binder. When using

561

an amorphous binder, the tablet tensile strength depended on granule size and deformability. In contrast,

562

granule strength was more important regarding tablet quality when using a brittle binder. However, this

563

was not the case for caffeine-containing blends, since these phenomena were dominated by the enhanced

564

compression properties of caffeine Form I. Therefore, it is important to gain knowledge about the behavior

565

of the blend during processing before the influence of the process parameters onto the granule and tablet

566

properties can be understood.

567

Based on the DOE models, optimal TS HMG parameter settings can be derived in order to obtain

568

granules having required characteristics to produce tablets with desired quality. In general, granules with

569

good flow properties (CI<15) and minimized amount of fines and oversized granules are preferred and tablets

570

should have a tensile strength >2MPa, tablet friability <1% and fast dissolution rate. For blend MPT/PEG,

571

the granule friability should be maximized which can be obtained by restricting the binder concentration.

572

In table 6, examples of such optimal parameter settings are given for the four studied formulations. The

573

calculated optima coincide with the findings of this research project. Table 6: Optimal factor settings for the four experimental designs based on the observations in current research project

MPT/PEG MPT/SLP CAF/PEG CAF/SLP

throughput (kg/h) max. (0.9) max. (0.8) max. (1.1) max. (1.0)

screw speed (rpm) min. (100) min. (200) max. (300) min. (200)

27

temperature ( ◦ C) > Tm < Tg > Tm Tg

% binder min. (5%) 10% (mean) max. (20%) 10% (mean)

574

Acknowledgment

575

Financial support for this research from the Agency for Innovation by Science and Technology (IWT

576

- Ph.D. fellowship Tinne Monteyne) is gratefully acknowledged. BASF is acknowledged for sending large

577

amounts of caffeine and Soluplusr .

578

579

580 581

5. References [1] J. Kowalski, O. Kalb, Y. M. Joshi, a. T. M. Serajuddin, Application of melt granulation technology to enhance stability of a moisture sensitive immediate-release drug product, International Journal of Pharmaceutics 381 (2009) 56–61.

582

[2] J. P. Lakshman, J. Kowalski, M. Vasanthavada, W.-Q. Tong, Y. M. Joshi, A. T. M. Serajuddin, Application of melt

583

granulation technology to enhance tabletting properties of poorly compactible high-dose drugs, Journal of Pharmaceutical

584

Sciences 100 (4) (2011) 1553–1565.

585 586 587 588 589 590 591 592

[3] C. Vervaet, J. P. Remon, Continuous granulation in the pharmaceutical industry, Chemical Engineering Science 60 (2005) 3949–3957. [4] K. Plumb, Major Challenges for the Pharmaceutical Industry Pharmaceutical Industry in the New, Pharmacia 070 (June) (2004) 1–32. [5] E. I. Keleb, A. Vermeire, C. Vervaet, J. P. Remon, Twin screw granulation as a simple and efficient tool for continuous wet granulation, International Journal of Pharmaceutics 273 (2004) 183–194. [6] B. Van Melkebeke, C. Vervaet, J. P. Remon, Validation of a continuous granulation process using a twin-screw extruder, International Journal of Pharmaceutics 356 (2008) 224–230.

593

[7] J. Vercruysse, D. C´ ordoba D´ıaz, E. Peeters, M. Fonteyne, U. Delaet, I. Van Assche, T. De Beer, J. P. Remon, C. Vervaet,

594

Continuous twin screw granulation: Influence of process variables on granule and tablet quality, European Journal of

595

Pharmaceutics and Biopharmaceutics 82 (1) (2012) 205–211.

596

[8] A. Kumar, J. Vercruysse, V. Vanhoorne, M. Toiviainen, P.-E. Panouillot, M. Juuti, C. Vervaet, J. P. Remon, K. V.

597

Gernaey, T. D. Beer, I. Nopens, Conceptual framework for model-based analysis of residence time distribution in twin-

598

screw granulation, European Journal of Pharmaceutical Sciences 71 (2015) 25–34.

599 600

[9] ICH Expert Working Group, Q8 Pharmaceutical Development, ICH Harmonised Tripartite Guideline 8 (August) (2009) 1–28.

601

[10] ICH Expert Working Group, Q9 Quality Risk Management (November) (2005) 1–23.

602

[11] ICH Expert Working Group, Q10 pharmaceutical quality system 22 (3) (2008) 177–181.

603

[12] G. A. V. Buskirk, S. Asotra, C. Balducci, P. Basu, G. DiDonato, A. Dorantes, W. M. Eickhoff, T. Ghosh, M. A. Gonz´ alez,

604

T. Henry, M. Howard, J. Kamm, S. Laurenz, R. MacKenzie, R. Mannion, P. K. Noonan, T. Ocheltree, U. Pai, R. P. Poska,

605

M. L. Putnam, R. R. Raghavan, C. Ruegger, E. S´ anchez, V. P. Shah, Z. J. Shao, R. Somma, V. Tammara, A. G. Thombre,

606

B. Thompson, R. J. Timko, S. Upadrashta, S. Vaithiyalingam, Best Practices for the Development, Scale-up, and Post-

607

approval Change Control of IR and MR Dosage Forms in the Current Quality-by-Design Paradigm, AAPS PharmSciTech

608

15 (3) (2014) 665–693.

28

609 610

[13] S. Jambhekar, Bioavailability and Granule Properties, in: Handbook of Pharmaceutical Granulation Technology, Third Edition, Drugs and the Pharmaceutical Sciences, CRC Press, 2009, pp. 487–497.

611

[14] V. Loureno, T. Herdling, G. Reich, J. C. Menezes, D. Lochmann, Combining microwave resonance technology to multi-

612

variate data analysis as a novel PAT tool to improve process understanding in fluid bed granulation, European Journal of

613 614 615

Pharmaceutics and Biopharmaceutics 78 (3) (2011) 513–521. [15] S. Verma, Y. Lan, R. Gokhale, D. J. Burgess, Quality by design approach to understand the process of nanosuspension preparation, International Journal of Pharmaceutics 377 (1-2) (2009) 185–198. doi:10.1016/j.ijpharm.2009.05.006.

616

[16] J. Huang, G. Kaul, C. Cai, R. Chatlapalli, P. Hernandez-Abad, K. Ghosh, A. Nagi, Quality by design case study:

617

An integrated multivariate approach to drug product and process development, International Journal of Pharmaceutics

618

382 (1-2) (2009) 23–32.

619 620

[17] T. Freeman, A. Birkmire, B. Armstrong, A QbD approach to continuous tablet manufacture, Procedia Engineering 102 (March) (2015) 443–449.

621

[18] T. Schæfer, Growth mechanisms in melt agglomeration in high shear mixers, Powder Technology 117 (2001) 68–82.

622

[19] T. Schaefer, C. Mathiesen, Melt pelletization in a high shear mixer. VIII. Effects of binder viscosity, International Journal

623 624 625

of Pharmaceutics 139 (1996) 125–138. [20] T. Monteyne, L. Heeze, The use of rheology to elucidate the granulation mechanisms of a miscible and immiscible system during continuous twin-screw melt granulation, International Journal of Pharmaceutics.

626

[21] V. Caron, L. Tajber, O. I. Corrigan, A. M. Healy, A comparison of spray drying and milling in the production of amorphous

627

dispersions of sulfathiazole/polyvinylpyrrolidone and sulfadimidine/ polyvinylpyrrolidone, Molecular Pharmaceutics 8 (2)

628

(2011) 532–542.

629

[22] V. Mazel, C. Delplace, V. Busignies, V. Faivre, P. Tchoreloff, N. Yagoubi, Polymorphic transformation of anhydrous

630

caffeine under compression and grinding: a re-evaluation., Drug development and industrial pharmacy 37 (7) (2011)

631

832–840.

632

[23] J. Vercruysse, A. Burggraeve, M. Fonteyne, P. Cappuyns, U. Delaet, I. Van Assche, T. De Beer, J. P. Remon, C. Vervaet,

633

Impact of screw configuration on the particle size distribution of granules produced by twin screw granulation, International

634

Journal of Pharmaceutics 479 (1) (2015) 171–180.

635

[24] S. Hubert, S. Briancon, A. Hedoux, Y. Guinet, L. Paccou, H. Fessi, F. Puel, Process induced transformations during

636

tablet manufacturing: Phase transition analysis of caffeine using DSC and low frequency micro-Raman spectroscopy,

637

International Journal of Pharmaceutics 420 (2011) 76–83.

638 639 640 641 642 643

[25] J. T. Fell, J. M. Newton, Determination of tablet strength by the diametral compression test, Journal of Pharmaceutical Sciences 59 (5) (1970) 688–691. [26] B. J. Ennis, Theory of granulation: an engineering perspective, Handbook of pharmaceutical granulation technology 3 (2010) 6–58. [27] S. M. Iveson, J. D. Litster, K. Hapgood, B. J. Ennis, Nucleation, growth and breakage phenomena in agitated wet granulation processes: A review, Powder Technology 117 (1-2) (2001) 3–39.

644

[28] D. Djuric, Soluplus, in: T. Reintjes (Ed.), Solubility Enhancement with BASF Pharma Polymers., 2011, pp. 67–72.

645

[29] A. S. El Hagrasy, J. R. Hennenkamp, M. D. Burke, J. J. Cartwright, J. D. Litster, Twin screw wet granulation: Influence

646 647

of formulation parameters on granule properties and growth behavior, Powder Technology 238 (2013) 108–115. [30] M. Fonteyne, A. L. Fussell, J. Vercruysse, C. Vervaet, J. P. Remon, C. Strachan, T. Rades, T. De Beer, Distribution of

29

648

binder in granules produced by means of twin screw granulation, International Journal of Pharmaceutics 462 (1-2) (2014)

649

8–10.

650

[31] T. Monteyne, L. Heeze, K. Old??rp, C. Vervaet, J. P. Remon, T. De Beer, Vibrational spectroscopy to support the link

651

between rheology and continuous twin-screw melt granulation on molecular level: A case study, European Journal of

652

Pharmaceutics and Biopharmaceutics 103 (2016) 127–135. doi:10.1016/j.ejpb.2016.03.030.

653

[32] T. Monteyne, L. Heeze, S. T. F. C. Mortier, K. Old¨ orp, R. Cardinaels, I. Nopens, C. Vervaet, J.-P. Remon, T. De

654

Beer, The use of Rheology Combined with Differential Scanning Calorimetry to Elucidate the Granulation Mechanism

655

of an Immiscible Formulation During Continuous Twin-Screw Melt Granulation, Pharmaceutical Researchdoi:10.1007/

656

s11095-016-1973-6.

657 658

[33] T. Vasconcelos, B. Sarmento, P. Costa, Solid dispersions as strategy to improve oral bioavailability of poor water soluble drugs, Drug Discovery Today 12 (23-24) (2007) 1068–1075.

659

[34] L. Farber, K. P. Hapgood, J. N. Michaels, X. Y. Fu, R. Meyer, M. A. Johnson, F. Li, Unified compaction curve model

660

for tensile strength of tablets made by roller compaction and direct compression, International Journal of Pharmaceutics

661

346 (1-2) (2008) 17–24.

662

[35] N. Passerini, B. Albertini, M. L. Gonz´ alez-Rodr´ıguez, C. Cavallari, L. Rodriguez, Preparation and characterisation of

663

ibuprofen-poloxamer 188 granules obtained by melt granulation, European Journal of Pharmaceutical Sciences 15 (2002)

664

71–78.

665 666

[36] D. Yang, R. Kulkarni, R. J. Behme, P. N. Kotiyan, Effect of the melt granulation technique on the dissolution characteristics of griseofulvin, International Journal of Pharmaceutics 329 (1-2) (2007) 72–80.

667

[37] A. Kumar, J. Vercruysse, M. Toiviainen, P. E. Panouillot, M. Juuti, V. Vanhoorne, C. Vervaet, J. P. Remon, K. V.

668

Gernaey, T. De Beer, I. Nopens, Mixing and transport during pharmaceutical twin-screw wet granulation: Experimental

669

analysis via chemical imaging, European Journal of Pharmaceutics and Biopharmaceutics 87 (2) (2014) 279–289.

670 671 672 673 674 675

[38] R. M. Dhenge, R. S. Fyles, J. J. Cartwright, D. G. Doughty, M. J. Hounslow, A. D. Salman, Twin screw wet granulation: Granule properties, Chemical Engineering Journal 164 (2-3) (2010) 322–329. [39] D. Djuric, P. Kleinebudde, Impact of screw elements on continuous granulation with a twin-screw extruder, Journal of Pharmaceutical Sciences 97 (11) (2008) 4934–4942. [40] D. Djuric, B. Van Melkebeke, P. Kleinebudde, J. P. Remon, C. Vervaet, Comparison of two twin-screw extruders for continuous granulation, European Journal of Pharmaceutics and Biopharmaceutics 71 (1) (2009) 155–160.

676

[41] K. T. Lee, A. Ingram, N. a. Rowson, Twin screw wet granulation: The study of a continuous twin screw granulator using

677

Positron Emission Particle Tracking (PEPT) technique, European Journal of Pharmaceutics and Biopharmaceutics 81 (3)

678

(2012) 666–673.

679 680 681 682 683 684 685 686

[42] Q. Li, V. Rudolph, B. Weigl, A. Earl, Interparticle van der Waals force in powder flowability and compactibility, International Journal of Pharmaceutics 280 (1-2) (2004) 77–93. [43] T. Durig, 4 Binders in Pharmaceutical Granulation, in: Handbook of Pharmaceutical Granulation Technology, CRC Press, 2009, pp. 78–97. [44] G. Dalziel, E. Nauka, F. Zhang, S. Kothari, M. Xie, Assessment of granulation technologies for an API with poor physical properties, Drug Development and Industrial Pharmacy (December 2011) (2012) 1–11. [45] C. Sun, M. W. Himmelspach, Reduced tabletability of roller compacted granules as a result of granule size enlargement, Journal of Pharmaceutical Sciences 95 (1) (2006) 200–206.

30

687 688 689 690 691 692 693 694

[46] T. Sebhatu, Relationships between the effective interparticulate contact area and the tensile strength of tablets of amorphous and crystalline lactose of varying particle size, European Journal of Pharmaceutical Sciences 8 (1999) 235–242. [47] S. J. Wu, C. Sun, Insensitivity of compaction properties of brittle granules to size enlargement by roller compaction, Journal of Pharmaceutical Sciences 96 (5) (2007) 1445–1450. [48] T. Yoshinari, R. T. Forbes, P. York, Y. Kawashima, The improved compaction properties of mannitol after a moistureinduced polymorphic transition, International Journal of Pharmaceutics 258 (1-2) (2003) 121–131. [49] G. Thoorens, F. Krier, B. Leclercq, B. Carlin, B. Evrard, Microcrystalline cellulose, a direct compression binder in a quality by design environment - A review, International Journal of Pharmaceutics 473 (1-2) (2014) 64–72.

31

Table 7: Influence of varying throughput and screw speed on the material residence time in the barrel (RT) and the screw fill (SF) and the influence thereof on the granule size. In general Higher binder concentration → lower granule and tablet friability

Miscibility

Influence of throughput depends on ease of binder distribution (binder distribution is required for coalescence)

Immiscibility

Higher process torque → less fines

High influence of throughput and barrel filling degree (nuclei are kneaded together)

Low screw speed → more needle-shaped granules

High throughput → Higher torque, larger granules, less fines, stronger granules, smaller span, improved flow properties, improved drug binder interaction Low screw speed → more spherical granules

Relative larger granule size (larger binding capacity)

Granulation T can exceed Tg /Tm of the binder

Relative smaller granule size

Low screw speed → less fines

Miscibility limits max. possible granulation T High Temperature → higher dissolution rate

High binder concentration → less fines

Soluplus

No effect of temperature

Effect of temperature depends on miscibility (CAF/SLP: higher T → larger, stronger granules with smaller span distribution (MPT/SLP → limited influence due to improved hydrogen bond formation)

PEG

Higher granule friability stimulates tablet tensile strength

MPT

Polymorphic transition at high binder concentration and/or high granulation T produced more dense granules, larger (more oversized) granules, more deformable granules → higher TS

Caffeine

High Temp → lower dissolution rate

More binder → lower dissolution rate

High screw speed → lower dissolution rate

High screw speed → lower tensile strength

Granule deformability stimulates tablet tensile strength (For caffeine: more binder or more caffeine Form I) (For MPT: more drug binder interaction at high barrel filling degree)

32

Immersion Drug Binder (ini+al step)

0 0

Immiscible drug-binder blend 0 0 0 0 4 Miscible drug-binder blend