Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


journal homepage: www.elsevier.com/locate/ejpb

Research paper

PAT for tableting: Inline monitoring of API and excipients via NIR
spectroscopy
Patrick R. Wahl a, Georg Fruhmann a, Stephan Sacher a, Gerhard Straka b, Sebastian Sowinski c,
Johannes G. Khinast a,d,
a
Research Center Pharmaceutical Engineering GmbH, Graz, Austria
b
G.L. Pharma GmbH, Lannach, Austria
c
Sentronic GmbH, Dresden, Germany
d
Institute for Process and Particle Engineering, Graz University of Technology, Graz, Austria

a r t i c l e i n f o a b s t r a c t

Article history: This paper describes the application and implementation of inline NIR spectroscopy in an industrial tablet
Received 4 February 2014 press. The content uniformity of a powder was analyzed via a NIR probe mounted on the feed frame. A
Accepted in revised form 27 March 2014 PLS model with four latent variables (R2 = 0.97, Q2 = 0.95) was developed for the Active Pharmaceutical
Available online 3 April 2014
Ingredient (API) and two main excipients (EX1, EX2), according to the mixture DoE. The RMSEP corre-
sponded to the relative errors of 2.7% for API, 1.7% for EX1 and 2.6% for EX2, compared to the nominal
Keywords: formulation. Transfer of the model, from the lab to an inline setup for manufacturing was achieved using
NIR spectroscopy
local centering. There was a good agreement between the results of inline NIR and drawn tablets ana-
Process Analytical Technology (PAT)
Feed frame
lyzed via UVVis. Notably, NIR indicated stochastic segregation behavior of the powder toward the
Tablet press end of the process, which was conrmed by the UVVis analysis. The outcome of our work was related
Content uniformity to the recently published Ph. Eur. chapter 2.9.47 Demonstration of uniformity of dosage units using large
Sample size sample sizes.
Calibration transfer 2014 Elsevier B.V. All rights reserved.

1. Introduction also be utilized in batch processes, even after their initial design
and approval, to optimize the existing processes. The most com-
Starting in 2004, the FDA [1] and ICH (Q8Q11) [2] have mon PAT analyzer in the manufacturing of solid dosage forms is
released a series of guidelines regarding Quality by Design (QbD) Near-infrared Spectroscopy (NIRS) and has been extensively
and Process Analytical Technology (PAT). The QbD framework described in the literature [47]. This paper addresses the use of
has led to an important paradigm change by giving pharmaceutical NIRS for tablet manufacturing.
companies the freedom to develop well-controlled and consis- NIR was used to monitor the content uniformity of intact tablets
tently optimized manufacturing processes. However, documenting [8], also in combination with physical properties, such as hardness
a science-based understanding of the process variability and the [913]. Further publications focused on the subsequent coating
Critical Process Parameters (CPPs) is required. Here, computer sim- process, where the thickness of the coating layer was determined
ulation can be benecial to provide a deeper understanding [3]. No [14] and its impact on the release prole was investigated [15].
additional approval is necessary for changes within a well-docu- In these publications, tablets were primarily analyzed manually,
mented range of CPPs (i.e., the design space) that satises the qual- resulting in limited sampling capabilities during routine manufac-
ity requirements of the product (i.e., the Critical Quality Attributes, turing. For real-time information about the process, inline methods
CQAs). are preferable. In the eld of coating, inline systems have been
The Process Analytical Technology (PAT) is key for the analysis reported for a pan coater [16] and a uid bed coater [17,18]. In
and control of manufacturing processes. Especially, continuous order to study content uniformity, NIR was used inline during ejec-
production concepts rely on real-time inline measurements of tion of tablets [19], at 6000 tablets per hour. Clearly, higher inspec-
the product quality and on respective control strategies. PAT can tion rates are needed for a 100% control. NIR Chemical Imaging
(NIR-CI) allows much higher throughputs, as multiple tablets can
Corresponding author. Research Center Pharmaceutical Engineering GmbH, be scanned simultaneously. Another approach that does not limit
Inffeldgasse 13/2, 8010 Graz, Austria. Tel.: +43 316 873 30403. the amount of tablets per hour (and is not limited to scanning only
E-mail address: khinast@tugraz.at (J.G. Khinast). the tablets surface) involves mounting a NIR sensor on the feed

http://dx.doi.org/10.1016/j.ejpb.2014.03.021
0939-6411/ 2014 Elsevier B.V. All rights reserved.
272 P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278

frame [20], i.e., to monitor the powder mixture similar to inline clutter followed by Net Analyte Pre-processing (NAP) [33]. A com-
monitoring of powder blenders [2125]. However, difculties con- parison between different methods of including pure component
cerning sample presentation in the feed frame arise, which are spectra and removing clutter were presented in [34], including
associated with the paddle wheel moving the powder. NAP, Improved Direct Calibration (IDC), Science Based Calibration
NIRS also allows analysis of the crystalline state of the API in (SBC) and Augmented Classical Least Squares Calibration (CLS).
tablets. Reectance NIR was used to study the polymorphic state Several transfer methods for handling differences between
of pharmaceutical materials, with similar prediction accuracy as spectrometers or differing environmental conditions were
transmission Raman [26]. Differentiation between different poly- described and evaluated in the literature, including Direct Stan-
morphic states, i.e., form A and B of Irbesatan, was achieved using dardization (DS), Piecewise Direct Standardization (PDS), Articial
NIR measurements [27]. Final tablets were analyzed via NIR-CI Neural Networks (ANN), wavelets, Orthogonal Signal Correction
techniques to determine the distribution of API and excipients. (OSC), Reverse Standardization (RS), slope and bias correction,
For API quantication, an augmented Multivariate Curve Resolu- Model Updating (MU) and Local Centering (LC) [3537]. The sug-
tion-Alternating Least Squares (MCR-ALS) method was applied gested methods for calibration transfer, if transfer samples are
[28,29]. After calculating the distribution map, further Multivariate available on both systems, are PDS [35], RS [36] or LC [37]. LC
Image Analysis (MIA) methods were applied to detect API clusters was preferred by Bergman et al. as the smallest number of transfer
in the tablets [29]. NIR-CI was also used in the coating distribution samples was needed and transfer was simple to perform [37]. If no
analysis [30] (see Table 1). transfer samples are available, MU was suggested [36].
An industrial application of PAT requires appropriate monitor- In this work, we present the use of inline NIR spectroscopy for
ing techniques and an accurate and robust model, i.e., a model that monitoring the content uniformity of tablets compacted in an
can handle differences in sample presentation between lab and industrial setting. The NIR probe was mounted on the feed frame
manufacturing, and calibration transfer strategies for dealing with of the tablet press. The spectra for the PLS model were collected
differences between spectrometers and different sample presenta- in the lab using premixes on a rotating plate. To enable predictions
tion. The simplest way to address spectral differences between lab- during manufacturing, the model was transferred via LC. We report
oratory-scale implementations and real process analyzers was here the use of a calculated transfer sample, consisting of the nom-
proposed by Blanco et al. [31]. Adding process spectra with inal mixture measured in the laboratory and an average of all pro-
observed deviations to the model spectra resulted in a more robust cess spectra during manufacturing. For validation, the NIR results
PLS model. In order to guarantee that process deviations are were compared with the ofine UVVis analysis of selected tablets.
included in the model, a method of monitoring the validity of cal- Both methods conrm segregation at the end of the process.
ibration over time is needed. Cogdill et al. suggested to monitor Finally, quality improvement opportunities by inline measure-
spectral features, i.e., Hotellings T2 and the residuals Q [32]. More ments are discussed and questions are raised concerning a new
advanced mathematical approaches for separating noise induced denition of the USP 905 Uniformity of Dosage Units for inline
by the process (clutter) were proposed, e.g., Noise Augmentation measurements.
(NA) of the calibration spectra with the spectral shapes from the
2. Materials and methods
Table 1
List of abbreviations. 2.1. Materials

Abbreviation Meaning
The tablet formulation consisted of a drug (API) load of about
ANN Articial Neural Networks 30% and two main excipients (EX1 and EX2), which in total
API Active Pharmaceutical Ingredient
accounted for about 65%. The other excipients had a total fraction
AV Acceptance Value
CLS Augmented Classical Least Squares of less than 5%. The composition of powder was simultaneously
CPP Critical Process Parameter monitored for API, EX1 and EX2 via NIR spectroscopy.
CQA Critical Quality Attribute
DoE Design of Experiments
DS Direct Standardization 2.2. Tablet press
EX Excipient
FDA U.S. Food and Drug Administration The compaction of tablets was carried out using a Kilian Synthe-
ICH International Conference on Harmonization
sis 500 (Kilian GmbH & Co. KG, Germany) rotary tablet press. For
IDC Improved Direct Calibration
LC Local Centering
routine manufacturing, the speed was set to 300,000 double con-
MCR-ALS Multivariate Curve Resolution-Alternating Least Squares cave tablets per hour. The level sensor in the powder inlet chute
MIA Multivariate Image Analysis halts the production once the lower acceptable level is reached.
MU Model Updating For research purposes, it was continued until the feed frame was
NA Noise Augmentation
empty and no more tablets could be pressed.
NAP Net Analyte Pre-processing
NIRS Near-infrared Spectroscopy
NIR-CI Near-infrared Chemical Imaging 2.3. NIR spectroscopy
OSC Orthogonal Signal Correction
PAT Process Analytical Technology
PCA Principle Component Analysis NIR spectra were collected in diffuse reection mode via the
PDS Piecewise Direct Standardization process spectrometer SentroPAT FO (Sentronic GmbH, Germany),
Ph. Eur. European Pharmacopoeia which is a diode array spectrometer with a spectral range of
PLS Partial Least Squares 11002200 nm and a resolution of 2 nm. To monitor the composi-
RMSECV Root-mean Square Error of Cross-validation
RMSEP Root-mean Square Error of Prediction
tion of the powder, a SentroProbe DR LS with a large spot size of
RS Reverse Standardization 6 mm was used. The probe had an internal light source and a
RSD Relative Standard Deviation unique internal micro-mechanical white reference directly behind
SBC Science Based Calibration the sample window, which allowed measuring the internal refer-
SNV Standard Normal Variate
ence spectrum whenever required, even during the process. Once
USP U.S. Pharmacopoeia
the internal reference standard was measured, any changes in
P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278 273

Fig. 1. Left: NIR probe mounted to observation window of feed frame. Right: Schematic drawing of feed frame and rotating dies with two NIR measurement positions, above
right and left paddle wheel. (For interpretation of the references to color in this gure legend, the reader is referred to the web version of this article.)

the signal path, including probe optics and glass ber transmission, Quantication was performed using a ve-point calibration curve
were calculated and used to correct the samples further measure- in the range of 25100 lg/ml with R2 = 0.9998.
ments. Drifts may be caused by changes in the environmental con-
ditions, such as temperature variations and vibration, or aging of
the light source and the resulting intensity loss. Correcting for 2.7. Chemometric model and mixture design
the drifts can signicantly improve the long-term stability of the
acquired spectra in routine operation during manufacturing. In order to correctly predict the composition of the powder, a
The software SentroSuite was used to control the spectrometer. Design of Experiments (DoE) approach was chosen to select the
The integration time was set to 0.02 s and an averaging window of mixtures that were used in the NIR model. The API, EX1 and EX2
150 spectra. The total acquisition time was 3 s per spectrum. An were varied 20% relative to their nominal value (e.g., 1523% for
internal drift correction was performed every 3 min, with an inte- a nominal value of 19%). The ternary mixtures were selected
gration time of 0.01 s, averaged over 100 spectra, to correct for according to a D-Optimal solution via eight experiments (numbers
heating of the probe head. 17 in Fig. 2), covering all corners and the center point of the mix-
ture space. At the center point, all constituents in the mixture had
2.4. Probe position in feed frame the nominal values. The remaining excipients (e.g., lubricants)
were kept constant throughout all mixtures. To achieve nearly
Collection of the powder spectra was performed in the feed equidistant steps in mass fraction for calibration, two further
frame of the tablet press. One inspection window was replaced experiments were conducted (numbers 8 and 9). Three additional
by an adapter for mounting of the NIR probe directly above the mixtures were chosen to validate the model (numbers 1012).
paddle wheel, where the powder was distributed into the tablet
dies. To ensure proper sample presentation, with the least varia-
tions and without window fouling, the probe had to be positioned
0.54
at the appropriate distance to the paddle wheel to monitor the
powder composition just before compaction. 0.52

2.5. Effective sample volume 0.50

0.48
The effective sample volume of NIR was estimated, based on the
following data: 6 mm spot size, 3 s integration time, 60 mm dis- 0.46
tance of the NIR probe window from the paddle wheel shaft, rotat-
ing at 25 rpm. The powder is assumed to move at 50% of the paddle 0.44
wheels circumferential velocity. During the production, around
15,600 spectra were collected. The sample mass per unit area 0.42

was roughly estimated to 15 mg/cm2 (corresponding penetration


0.40
depth = 0.5 mm), for a spectral range between 1400 nm and
2200 nm, according to [38]. Thus, each spectrum sampled around 0.38
0.13 unit doses, totaling to a sample size of n = 2028.
0.36
2.6. UVVis reference analytics

Reference measurements of the API content were conducted Excipient 2


using the Perkin Elmer LAMBDA 950 UVVis spectrometer. The
tablets were weighed prior to UVVis analysis to correct for weight Fig. 2. Ternary mixture design for NIR calibration measurements. Points 17 (blue)
are according to a D-Optimal design; points 8 and 9 (black) are chosen to achieve
variations. After pre-ltering through a 0.2 lm cellulose nitrate l- nearly equidistant steps in content for each component; points 1012 (red) are for
ter (Sartorius, Germany), the tablets were dissolved in 20 ml EtOH validation purposes. (For interpretation of the references to color in this gure
and diluted 1:50 with puried water (MicroPure, TKA, Germany). legend, the reader is referred to the web version of this article.)
274 P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278

Mixtures of 15 g each were prepared and mixed in a Turbula 3. Results and discussion
T2F tumbler blender (Maschinenfabrik Willy A. Bachofen AG,
Switzerland) for 5 min at 60 Hz, followed by adding the lubricant 3.1. PCA analysis to detect process irregularities
and mixing for additional 2 min. To avoid segregation after
mixing, the powder was carefully distributed on a rotary plate An industrial tableting process with a batch process time of
rotating at a speed of around 30 rpm (i.e., 2 s per revolution). around 13 h was monitored via NIR in the feed frame, as described
Further details of the procedure can be found in Scheibelhofer above. Throughout the process, a PCA of the original spectra was
et al. [23]. performed to check for any major deviations. The rst principle
With the collected spectra, a chemometric model was built with component (98% explained variance) captured the baseline shift
Simca 13 (Umetrics, Sweden). The spectra were pretreated with of the spectra toward the process end. Toward the end of the pro-
SNV (12002160 nm), followed by rst-order derivate with Sav- cess, when the feed frame emptied, the distance between the probe
itzkyGolay smoothing (second-order polynomial, kernel: 23 head and the powder surface increased, resulting in less reected
points) in the same wavelength region (see Fig. 3). A PLS model light. The PC1 vs. PC2 for the entire 13-h process are plotted in
(12142146 nm, four latent variables) was applied, with a predic- Fig. 5, left. While comparably minor deviations were observed dur-
tive capability of R2 = 0.97, Q2 = 0.95. The RMSEP and RMSECV val- ing the process, strong outliers became apparent toward the pro-
ues, of prediction and cross-validation, respectively, were cess end. In Fig. 5, right, the second principle component is
calculated as relative error in relation to the nominal formulation. shown (2% explained variance), which correlated best with the
The calculated values of RMSEP (RMSECV) were 2.7% (4.4%) for API, API content according to the loadings. The loadings of PC2 showed
1.7% (2.4%) for EX1 and 2.6% (4.5%) for EX2. no noise, despite the low explained variance. The process
Validation experiments (Fig. 2, numbers 1012) exhibited rela- stopped at around 400 s (Fig. 5(a)), which was triggered by the
tive RMSEP values for API: 2.0%, EX1: 0.69% and EX2: 2.3%, respec- level sensor in the powder inlet chute between the hopper and
tively, indicating a robust model (see Fig. 4). the feed frame. Shortly thereafter, the tablet press was restarted
To use the model for the monitoring of a real tableting pro- and continued until no more powder was left in the feed frame
cess, further effects had to be considered. The paddle wheel of (Fig. 5(c)). Just before the end of the process, pronounced devia-
the tablet press, which distributes the powder into the dies, tions in PC2 were observed (Fig. 5(b)), indicating an excess API con-
passes directly under the probe, causing agitation of the powder tent in the mixture. Since this could have led to overly potent
and possibly affecting the spectrum. A LC transformation, neces- tablets, further investigations were performed.
sary to correct for these effects, was based on assuming a correct
overall composition of the powder for tableting, which was rea-
3.2. Conrmation of deviations with UVVis
sonable for an industrial process. The transfer sample for LC was
calculated instead of measured. The average spectrum of the
To conrm the ndings of the PCA analysis, the tablets were
process Aprocess was calculated, with all major deviations
sampled and analyzed by means of UVVis spectroscopy. At two
excluded (e.g., start, end, stop). This averaged spectrum of the
points during the process (after around 4 h and 9 h), samples con-
process had to be identical to the average spectrum of the center
sisting of 5 tablets were taken and analyzed for inter-tablet uctu-
point Acenter point of the mixture DoE, which had the same compo-
ations. The Relative Standard Deviation (RSD) was found to be
sition as the formulation. After normalizing the minimum and
RSDsample = 3%. Additionally, 13 samples were drawn throughout
maximum absorbance values of the spectra between zero and
the process at intervals of about 1 h (Fig. 6). For the process, a
one (zero-to-one normalization, 12002160 nm), the centering
RSDprocess = 5.4% was determined, which was only 80% higher than
was performed using the equation:
a single sample and which suggested a stable process with minor,
well-controlled variations over time. A discussion of the API con-
Ai;centered Ai;process  Aprocess Acenter point
tent tolerances with regard to the inline monitoring is presented
below.
where Ai,process is a spectrum taken in the feed frame and Ai,centered is
In order to resolve the end of the process, regular samples were
the centered spectrum. The PLS model was applied to these cor-
taken in 30-s intervals starting at the point of time where the pro-
rected spectra to accurately predict the powder composition in
cess was re-started. The UVVis analysis of samples from three
the feed frame.
batches conrmed the PCA ndings (Fig. 6). Variations in the API
content of 12% (Fig. 6, left) to 30% (Fig. 6, right) of the label claim
were identied, which are likely due to (stochastic) segregation
0.08
effects in hopper feeding powder to the feed frame. Many param-
Absorbance (SNV, 1 derivate)

eters inuence the segregation tendency during discharge of a


0.06 hopper, including particle diameter ratio and nes mass fraction,
the hopper cross-sectional shape and hopper angle [39]. The vol-
st

0.04 ume mean particle size of the API was 140 lm, which was signi-
cantly larger compared to the non-granulated excipients. Size
0.02
segregation during discharge of the hopper, leading to an accumu-
0 lation of large API crystals toward the process end [40], could have
occurred. The vibrations of the tablet press might have induced
0.02 further segregation [41,42]. To conrm reproducibility, the batch
with the most pronounced deviations was analyzed in triplicate
0.04 (Fig. 6, right).
1200 1300 1400 1500 1600 1700 1800 1900 2000 2100
Wavelength [nm]
3.3. Inline monitoring of the powder composition
Fig. 3. Model spectra after pretreatment with SNV and rst order derivative.
Colored according to mixtures (19) of the DoE. (For interpretation of the references
to color in this gure legend, the reader is referred to the web version of this Based on the conrmed variations, a PLS model to predict the
article.) API and two main excipients was developed and used to analyze
P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278 275

Fig. 4. Observed vs. predicted plots for PLS model for API, EX1 and EX2. Symbols: circles for calibration and diamonds for validation experiments.

Fig. 5. Left: PC1 vs. PC2 of the entire production process. Only a small number of measurements deviate strongly. Right: Recognizable process events during the last ten
minutes of the process: (a) process stop, (b) possibly too much API and (c) powder emptying. The spectra (b) and (c) correspond to the observed deviations on the left side.

Fig. 6. Signicant deviations in API content (up until +30% of label claim) for three batches toward the end of the process.

several production runs, one of which is discussed below. Pre-tests stated relative to the nominal content of the formulation. Note that
were performed to select the best measurement position above the the API excess (blue), e.g., occurring between 10 and 20 min, is
left or right paddle wheel (see Fig. 1). Finally, all measurements compensated by the reduction in only one excipient (red, EX2),
were performed above the left paddle wheel for two reasons: First, which may be a useful observation with regard to future improve-
the time interval between taking spectra and tableting is shorter ments to the formulation. Additionally, out-of-spec tablets could
and subsequent (de-)mixing effects are minimized. Second, since possibly be identied selected and removed, which would drasti-
the sample presentation was better, with less (long-term) varia- cally increase the overall quality. Fig. 7, right, presents a compari-
tions in the captured spectra (not shown), the left position was son of inline (NIR) and ofine (UVVis) methods. A time delay of on
selected. These variations may be due to the way the powder is average 30 s between NIR and UVVis data was observed and cor-
lled into the die holes: the left paddle transported the powder rected for in the gure for comparison purposes. During the time-
to the turret and into the die holes, while the right paddle mainly frame of 30 s, further (de-)mixing could have occurred, but clearly
removes and recycles the superuous powder, which is later trans- there is strong correlation between inline and ofine results. This
ported back to the left paddle and re-distributed into the die holes. conrms that the composition of the tablets can be predicted via
Since the powder at the right paddle wheel is more mechanically NIR monitoring of the powder.
stressed and experiences higher shear forces, more nes could
appear due to attrition and the powder could potentially segregate. 3.4. Quality improvement opportunities by inline analysis
This effect, alongside with the segregation in the hopper, might
have been root causes for content deviations toward the process According to the U.S. Pharmacopoeia (USP 905 Uniformity of
end. Dosage Units), a batch of tablets is considered in-spec with
A typical plot of the three component concentrations during the regard to content uniformity, if the Acceptance Value (AV) of the
last hour of the process is shown in Fig. 7, left. All components are sampled tablets is smaller than 15. The AV is calculated as
276 P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278

Fig. 7. Left: Plot of the entire formulation over time, with strong API deviations toward the end of the process. Right: Comparison of UVVis and NIR results at the process end
(UVVis data shifted by 30 s to compensate for the time delays between the NIR measurement and sampling). (For interpretation of the references to color in this gure
legend, the reader is referred to the web version of this article.)

AV jM  Xj ks, with the reference value M, the mean API content on the number of samples. The number of samples can be trans-
of the analyzed tablets X, the acceptability constant k and the RSD s. lated to a fraction of deviating samples compared to overall drawn
Assuming that the deviations in the mean API content are less than samples, as shown in Fig. 8. Notably the fraction of samples, which
1.5%, the formula can be simplied as AV = ks, with k = 2.4 for n = 10 are allowed to deviate above L2, increases with sample size, as the
tablets and k = 2.0 for n = 30 tablets analyzed. For the analyzed batch statistical uncertainty decreases. In contrast, the fraction of sam-
(Fig. 6, right, and Fig. 7), the acceptance value for the reference ples deviation more than L1 is slightly decreasing. In our experi-
samples analyzed via UVVis (RSDprocess,UVVis = 5.4%, 10 tablets: ments, an estimated sample size of n = 2028 was analyzed.
k = 2.4) was AV = 2.4 * 5.3 = 12.72 and for the inline NIR analysis According to Fig. 8, the acceptable number of dosage units with a
(RSDNIR = 3.3%, >30 unit doses: k = 2.0) it was AV = 2.4 * 3.3 = 6.6. content deviating more than L2 is 18 [44], or 0.89% of the samples
Hence, the batch was in-spec according to the reference and the drawn. Despite the observed deviations toward the process end, all
inline measurements. monitored batches are in-spec.
Moreover, no tablet should deviate from the label claim by
more than 25%. Thus, the analyzed batches could fail if a tablet 4. Conclusion
was drawn at the very end of the process, when the API content
was above 125% of the label claim. This critical timeframe of about A staged approach for optimizing an existing tablet production
30 s constituted only 0.064% of the overall process time of 13 h. was proposed. Inline NIR spectroscopy indicated that the end of the
The chance of sampling at least one of these tablets was calculated process was critical: a signicant peak in the API content was
by p = 1  (1  0.00064)n with values of 0.64% for n = 10 tablets observed, which was probably caused by segregation effects. Fur-
and 1.91% for n = 30 tablets. Thus, even assuming a totally random ther comparison of drawn samples and inline predicted powder
sampling, it would be unlikely that classical thief sampling composition conrmed the ndings, and corrective actions were
schemes of reference tablets would detect high frequency process suggested.
upsets. However, inline methods (e.g., spectroscopy) can resolve In our case, inline analysis proved to have distinct advantages
such events. because it created a more complete picture of the process than
One of the main concerns using inline methods is the USP 905 the AV, which is only a single statistical number. Since sampling
criteria that no tablet is allowed to deviate more than 25% from a few tablets may not provide sufcient information about process
the nominal composition [43]. Fast measurements result in a large events that lead to product quality deviations, conventional pro-
number of samples, thus strongly increasing the probability of at cess and quality control may fail to determine the signicance
least on sample to fail the criteria. The recently published Euro- and effects of those critical events. Process analytical technologies,
pean Pharmacopoeia chapter 2.9.47 Demonstration of uniformity such as NIRS, are key enablers of the next step in quality and pro-
of dosage units using large sample sizes addresses this problem cess understanding in the pharmaceutical industry.
[44]. According to the chapter, a certain number of samples are The denition of Ph. Eur. chapter 2.9.47 refers to the number of
allowed to deviate more than L1 = 15% and L2 = 25%, depending drawn samples. For spectroscopic measurements, calculating this

Fig. 8. The fraction of drawn samples, which are allowed to deviate above L1 (left) and L2 (right), according to data from [44].
P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278 277

number is not simple. The number of drawn samples depends on [14] J.J. Moes, M.M. Ruijken, E. Gout, H.W. Frijlink, M.I. Ugwoke, Application of
process analytical technology in tablet process development using NIR
the effective sample volume. This in turn directly depends on the
spectroscopy: blend uniformity, content uniformity and coating thickness
samples mass per unit area or the effective sample thickness, as measurements, Int. J. Pharm. 357 (2008) 108118.
well as the movement of the powder in front of the sensor. Exper- [15] S.H. Tabasi, R. Fahmy, D. Bensley, C. OBrien, S.W. Hoag, Quality by design, Part
imental determination of these parameters, especially for agitated II: Application of NIR spectroscopy to monitor the coating process for a
pharmaceutical sustained release product, J. Pharm. Sci. 97 (2008) 40524066.
powder, is delicate. The authors expect notable differences [16] M.-J. Lee, D.-Y. Seo, H.-E. Lee, I.-C. Wang, W.-S. Kim, M.-Y. Jeong, et al., In line
between how companies measure or dene these parameters. NIR quantication of lm thickness on pharmaceutical pellets during a uid
Hence, the exact number of sampled tablets by spectroscopy is bed coating process, Int. J. Pharm. 403 (2011) 6672.
[17] J.D. Prez-Ramos, W.P. Findlay, G. Peck, K.R. Morris, Quantitative analysis of
nearly impossible to determine. A clear denition of the sample lm coating in a pan coater based on in-line sensor measurements, AAPS
volume is still missing and should be given by regulatory authori- PharmSciTech 6 (2005). E127E136.
ties. By using inline measurements, out-of-spec materials can be [18] M. Andersson, S. Folestad, J. Gottfries, M.O. Johansson, M. Josefson, K.-G.
Wahlund, Quantitative analysis of lm coating in a uidized bed process by in-
detected in real time and thus can be immediately removed from line NIR spectrometry and multivariate batch calibration, Anal. Chem. 72
the batch. Thus, it should be allowed to remove these out-of-spec (2000) 20992108.
tablets from the sample statistics. [19] A.D. Karande, P.W.S. Heng, C.V. Liew, In-line quantication of micronized drug
and excipients in tablets by near infrared (NIR) spectroscopy: real time
Overall, inline analysis could lead to signicant improvements monitoring of tabletting process, Int. J. Pharm. 396 (2010) 6374.
in product quality. This discussion leads to the important question: [20] H.W. Ward, D.O. Blackwood, M. Polizzi, H. Clarke, Monitoring blend potency in
Does drawing a handful of tablets still constitute the state of the a tablet press feed frame using near infrared spectroscopy, J. Pharm. Biomed.
Anal. 80 (2013) 1823.
art for quality control or would it be wise to demand inline
[21] S.S. Sekulic, H.W. Ward II, D.R. Brannegan, E.D. Stanley, C.L. Evans, S.T.
measurements? Sciavolino, et al., On-line monitoring of powder blend homogeneity by near-
infrared spectroscopy, Anal. Chem. 68 (1996) 509513.
[22] O. Scheibelhofer, N. Balak, P.R. Wahl, D.M. Koller, B.J. Glasser, J.G. Khinast,
Acknowledgements Monitoring blending of pharmaceutical powders with multipoint NIR
spectroscopy, AAPS PharmSciTech 14 (2013). E234E244.
[23] O. Scheibelhofer, N. Balak, D.M. Koller, J.G. Khinast, Spatially resolved
This work has been funded by the Austrian COMET Program monitoring of powder mixing processes via multiple NIR-probes, Powder
under the auspices of the Austrian Federal Ministry of Transport, Technol. 243 (2013) 161170.
Innovation and Technology (bmvit), the Austrian Federal Ministry [24] Y. Sulub, B. Wabuyele, P. Gargiulo, J. Pazdan, J. Cheney, J. Berry, et al., Real-time
on-line blend uniformity monitoring using near-infrared reectance
of Economy, Family and Youth (bmwfj) and by the State of Styria spectrometry: a noninvasive off-line calibration approach, J. Pharm. Biomed.
(Styrian Funding Agency SFG). COMET is managed by the Austrian Anal. 49 (2009) 4854.
Research Promotion Agency FFG. The authors would like to thank [25] C.V. Liew, A.D. Karande, P.W.S. Heng, In-line quantication of drug and
excipients in cohesive powder blends by near infrared spectroscopy, Int. J.
GL Pharma for supplying the materials and providing the opportu- Pharm. 386 (2010) 138148.
nity to work together with their experienced team. [26] M.C. Hennigan, A.G. Ryder, Quantitative polymorph contaminant analysis in
tablets using Raman and near infra-red spectroscopies, J. Pharm. Biomed. Anal.
72 (2013) 163171.
References [27] D. Pan, G. Crull, S. Yin, J. Grosso, Low level drug product API form analysis
avalide tablet NIR quantitative method development and robustness
challenges, J. Pharm. Biomed. Anal. 89 (2014) 268275.
[1] U.S. Food and Drug Administration, Guidance for Industry. PAT A Framework
[28] J.M. Amigo, C. Ravn, Direct quantication and distribution assessment of major
for Innovative Pharmaceutical Development, Manufacturing, and Quality
and minor components in pharmaceutical tablets by NIR-chemical imaging,
Assurance, US Department of Health, Rockville, MD, 2004.
Eur. J. Pharm. Sci. 37 (2009) 7682.
[2] ICH, ICH Q8(R2) (Pharmaceutical Development), ICH Q9 (Quality Risk
[29] J.M. Prats-Montalbn, J.I. Jerez-Rozo, R.J. Romaach, A. Ferrer, MIA and NIR
Management), ICH Q10 (Pharmaceutical Quality System) and ICH Q11
chemical imaging for pharmaceutical product characterization, Chemom.
(Development and Manufacture of Drug Substances).
Intell. Lab. Syst. 117 (2012) 240249.
[3] S. Adam, D. Suzzi, C. Radeke, J.G. Khinast, An integrated Quality by Design
[30] A. Palou, J. Cruz, M. Blanco, J. Toms, J. de los Ros, M. Alcal, Determination of
(QbD) approach towards design space denition of a blending unit operation
drug, excipients and coating distribution in pharmaceutical tablets using NIR-
by Discrete Element Method (DEM) simulation, Eur. J. Pharm. Sci. 42 (2011)
CI, J. Pharm. Anal. 2 (2012) 9097.
106115.
[31] M. Blanco, R. Cueva-Mestanza, A. Peguero, NIR analysis of pharmaceutical
[4] M. Blanco, J. Coello, H. Iturriaga, S. Maspoch, C. De Pezuela, Near-infrared
samples without reference data: improving the calibration, Talanta 85 (2011)
spectroscopy in the pharmaceutical industry, Analyst 123 (1998) 135150.
22182225.
[5] T. De Beer, A. Burggraeve, M. Fonteyne, L. Saerens, J.P. Remon, C. Vervaet, Near
[32] R.P. Cogdill, C.A. Anderson, J.K. Drennen, Process analytical technology case
infrared and Raman spectroscopy for the in-process monitoring of
study, Part III: Calibration monitoring and transfer, AAPS PharmSciTech 6
pharmaceutical production processes, Int. J. Pharm. 417 (2011) 3247.
(2005). E284E297.
[6] J. Luypaert, D.L. Massart, Y. Vander Heyden, Near-infrared spectroscopy
[33] S. Pieters, W. Saeys, T. Van den Kerkhof, M. Goodarzi, M. Hellings, T. De Beer,
applications in pharmaceutical analysis, Talanta 72 (2007) 865883.
et al., Robust calibrations on reduced sample sets for API content prediction in
[7] G. Reich, Near-infrared spectroscopy and imaging: basic principles and
tablets: denition of a cost-effective NIR model development strategy, Anal.
pharmaceutical applications, Adv. Drug Deliv. Rev. 57 (2005) 11091143.
Chim. Acta 761 (2013) 6270.
[8] W. Li, L. Bagnol, M. Berman, R.A. Chiarella, M. Gerber, Applications of NIR in
[34] S. Sharma, M. Goodarzi, L. Wynants, H. Ramon, W. Saeys, Efcient use of pure
early stage formulation development. Part II. Content uniformity evaluation of
component and interferent spectra in multivariate calibration, Anal. Chim.
low dose tablets by principal component analysis, Int. J. Pharm. 380 (2009) 49
Acta 778 (2013) 1523.
54.
[35] R.N. Feudale, N.A. Woody, H. Tan, A.J. Myles, S.D. Brown, J. Ferr, Transfer of
[9] P. Schoenmakers, R. Smits, A. Townshend, M. Blanco, M. Alcal, Content
multivariate calibration models: a review, Chemom. Intell. Lab. Syst. 64 (2002)
uniformity and tablet hardness testing of intact pharmaceutical tablets by near
181192.
infrared spectroscopy, Anal. Chim. Acta 557 (2006) 353359.
[36] C.F. Pereira, M.F. Pimentel, R.K.H. Galvo, F.A. Honorato, L. Stragevitch, M.N.
[10] M. Blanco, M. Alcal, J.M. Gonzlez, E. Torras, A process analytical technology
Martins, A comparative study of calibration transfer methods for
approach based on near infrared spectroscopy: tablet hardness, content
determination of gasoline quality parameters in three different near infrared
uniformity, and dissolution test measurements of intact tablets, J. Pharm. Sci.
spectrometers, Anal. Chim. Acta 611 (2008) 4147.
95 (2006) 21372144.
[37] E.-L. Bergman, H. Brage, M. Josefson, O. Svensson, A. Sparn, Transfer of NIR
[11] R.P. Cogdill, C.A. Anderson, M. Delgado-Lopez, D. Molseed, R. Chisholm, R.
calibrations for pharmaceutical formulations between different instruments, J.
Bolton, et al., Process analytical technology case study, Part I: Feasibility
Pharm. Biomed. Anal. 41 (2006) 8998.
studies for quantitative near-infrared method development, AAPS
[38] O. Berntsson, T. Burger, S. Folestad, L.G. Danielsson, J. Kuhn, J. Fricke, Effective
PharmSciTech 6 (2005). pp. E262E272.
sample size in diffuse reectance near-IR spectrometry, Anal. Chem. 71 (1999)
[12] R.P. Cogdill, C.A. Anderson, M. Delgado, R. Chisholm, R. Bolton, T. Herkert, et al.,
617623.
Process analytical technology case study, Part II: Development and validation
[39] W.R. Ketterhagen, J.S. Curtis, C.R. Wassgren, A. Kong, P.J. Narayan, B.C.
of quantitative near-infrared calibrations in support of a process analytical
Hancock, Granular segregation in discharging cylindrical hoppers: a discrete
technology application for real-time release, AAPS PharmSciTech 6 (2005). pp.
element and experimental study, Chem. Eng. Sci. 62 (2007) 64236439.
E273E283.
[40] N. Standish, Studies of size segregation in lling and emptying a hopper,
[13] S.H. Tabasi, R. Fahmy, D. Bensley, C. OBrien, S.W. Hoag, Quality by design, Part
Powder Technol. 45 (1985) 4356.
I: Application of NIR spectroscopy to monitor tablet manufacturing process, J.
Pharm. Sci. 97 (2008) 40404051.
278 P.R. Wahl et al. / European Journal of Pharmaceutics and Biopharmaceutics 87 (2014) 271278

[41] A.D. Rosato, D.L. Blackmore, N. Zhang, Y. Lan, A perspective on vibration- [43] O. Holte, M. Horvat, Evaluation of uniformity of dosage units using large
induced size segregation of granular materials, Chem. Eng. Sci. 57 (2002) 265 sample sizes, Pharmeuropa 23 (2011) 286293.
275. [44] General Chapter 2.9.47 Demonstration of Uniformity of Dosage Units using
[42] D. Huerta, J. Ruiz-Surez, Vibration-induced granular segregation: a Large Sample Sizes, Eur. Pharmacopoeia, Council of Europe, Strasbourg, France.
phenomenon driven by three mechanisms, Phys. Rev. Lett. 92 (2004) 114301.

You might also like