Sie sind auf Seite 1von 11

J Pharm Innov (2014) 9:291301

DOI 10.1007/s12247-014-9194-1

RESEARCH ARTICLE

Near Infrared Method Development for a Continuous


Manufacturing Blending Process
Yleana M. Coln & Miguel A. Florian & David Acevedo &
Rafael Mndez & Rodolfo J. Romaach

Published online: 30 August 2014


# Springer Science+Business Media New York 2014

Abstract
Purpose This study describes the validation of a near infrared
spectroscopic method for monitoring a continuous
manufacturing system. The achievement of this goal requires
determining the near infrared (NIR) methods accuracy, precision, obtaining an estimate of the sample volume analyzed,
and the frequency with which measurements should be
obtained.
Methods Five calibration blends were prepared spanning a
concentration range from 7 to 13 % w/w ibuprofen to prepare
a partial least squares (PLS) calibration model for a five
component formulation. NIR spectra were obtained after powders passed through feeders and a custom-made tumble mixer.
The calibration models precision and accuracy were determined with the prediction of three validation blends.
Results NIR concentration predictions obtained from spectra
collected during the validation runs showed relative standard
errors of predictions of 2.8 % at target concentration with
standard deviations NMT 0.2 % w/w. At certain time points,
blend samples coming out of the blender were collected for
UV analysis. Results from the NIR and UV analysis were
comparable with the largest difference being 0.36 % w/w
between the methods. A continuous blending process was
monitored for 3 min after steady-state conditions were
achieved. All individual predictions were within 3 standard
deviations of the average reference value. The standard deviation for the NIR concentration predictions was 0.49 % w/w.
Conclusion The use of the standard normal variate (SNV)
transform significantly reduced the effect of differences in
Y. M. Coln : R. J. Romaach (*)
Department of Chemistry, University of Puerto Rico at Mayagez,
PO Box 9000, Mayagez Campus, Mayagez 00682, Puerto Rico
e-mail: rodolfoj.romanach@upr.edu
M. A. Florian : D. Acevedo : R. Mndez
Department of Chemical Engineering, University of Puerto Rico at
Mayagez, Mayagez, Puerto Rico

powder flow on the NIR predictions. The use of variograms


provided valuable insight into the frequency of measurements needed for the continuous manufacturing system.
Additional research is needed to investigate the differences
observed in the NIR and UV results for the continuous
manufacturing run.
Keywords Continuous powder mixing . Near infrared
spectroscopy . Validation . Sampling . Pharmaceuticals .
Particulate processes . Theory of sampling . In-line
quantification . Calibration

Introduction
Current pharmaceutical manufacturing processes primarily
involve batch processes. These batch processes require that
considerable development efforts be dedicated to product
scale-up and transference of the process from development
to manufacturing facilities [14]. The use of a continuous
manufacturing process, as described in this work, will eliminate scale-up efforts as the same mixing unit used in development could be used in product manufacture.
The continuous manufacturing process described in this
paper uses a small custom-made mixer that offers significant
flexibility at a maximum of 80 kg/h at 70 rpm [5]. The
excipients and active pharmaceutical ingredient (API) are
added to the mixer through volumetric feeders. The size of a
lot will not be restricted by the manufacturing equipment
available at a pharmaceutical site. Lot size will be adjusted
by the amount of time employed during manufacture. Thus,
the system will be used for a certain time to fulfill a customers
order of 100,000 tablets and for a longer period for a 500,000
tablet order. The continuous manufacturing system is also a
very compact system, thereby reducing product hand-offs and
transfers, reducing the potential for product cross-

292

contamination and simplifying cleaning validation efforts.


Due to the small size of continuous manufacturing systems,
product isolation could be facilitated for potent drugs, although the design presented in this study is not designed for
these purposes. The small size of continuous manufacturing
mixers is also an advantage in early product development
when supplies of a new API are often limited. The increased
flexibility may be achieved within a smaller manufacturing
area leading to considerable savings in energy and site
maintenance.
Near infrared (NIR) spectroscopy has been extensively
used for monitoring batch processes [68], but this work
describes the monitoring of a continuous mixing process with
NIR spectroscopy. The NIR methods for batch manufacturing
have been used to evaluate the endpoint of the mixing process
[9, 10]. The NIR methods for continuous blending need to
monitor the variation in the mixing after steady state is
achieved [11, 12]. The evaluation of method precision is
extremely important to discern between the variation from
the mixing process and the variation associated to the NIR
method that is monitoring the blending process. Thus, in this
work, the precision and accuracy of the NIR method are
thoroughly evaluated. A five-component blend is analyzed
and significant efforts to incorporate previous knowledge
from this field which has shown the importance of carefully
constructing calibration sets capable of predicting the future
manufacturing process [12].

Materials and Methods


Materials
Ibuprofen (90 grade, BASF Corporation) was chosen as a
representative cohesive API, lactose monohydrate (Tablettose
70, Mutchler Inc., NF, EP, JP) and microcrystalline cellulose
Type 102 (JRS Pharma, PhEur, JP, USP, NF) were chosen as
main excipients and accounted for roughly 90 % of the target
blend. Colloidal silicon dioxide (99.0100.8 % (w/w), Cabot
Corporation) and magnesium stearate (Mallinckrodt Inc., NF)
were also included as minor excipients to enhance ease of
flow and final product parameters.
Manufacturing Setup
Figure 1 illustrates the experimental continuous manufacturing setup used for these experiments (model development,
validation runs, and continuous process). Pilot-scale feeders
allowed flow rate control of powder to the mixer. The NIR
instrument was placed on a holder over the conveyor belt.
Preblended formulations were placed in a Gericke feeder
initially filled up to 80 % of its maximum capacity and
operated in volumetric mode (the screw operated at constant

J Pharm Innov (2014) 9:291301

speed, 76.5 kg/h). For the continuous blending process run,


API preblend was placed in feeder #1 and the excipient
preblend in feeder #2 (the screw operated at constant speed,
9.96 and 66.5 kg/h, respectively).

NIR Spectral Acquisition


A Control Development, Inc. (CDI) spectrometer series 1402
NIR analyzer (South Bend, IN) governed by CDI Spec32 data
acquisition package version 1.7.1.3 was used to obtain the
NIR spectra. This spectrometer is equipped with an indium
gallium arsenide (InGaAs) diode array that is
thermoelectrically cooled and has 256 elements covering a
spectral response from 905 to 1,681 nm and includes a diffuse
reflectance measurement probe that illuminates a 2.5-cm diameter. The NIR instrument was placed approximately 1 cm
over the sample. The systems integration time was set at
6.6 ms per scan. A total of 36 spectra were averaged.
Reference spectra were obtained with a CDH 50
Standard White disk, made with Albrillon, an organic
microcrystalline fluorinated polymer that provides a
strong reflectance. This disk was also used to characterize the spectral noise. The spectrum of this material was
used for both background and sample spectra, and the
ratio of the spectra was used to estimate the instrument
noise. The standard deviation of the mean signal among
days ranged from 1.7 to 2.0104. Spectra were acquired during five consecutive days. The system was
turned off and on for periods of 20 min each day. A
15-min wait time for lamp stability was employed as
recommended by the manufacturer prior to collecting
spectra. This result represents the minimum standard
deviation estimated to be observed throughout the
experiments.
The sample volume analyzed by the NIR was estimated by
placing layers of different width of sample on top of an acrylic
surface. As the sample thickness on top of the acrylic surface
was decreased, the distinct absorption bands of the acrylic
surface around 1,331 and 1,344 nm were observed. These
characteristic bands were first observed at approximately
0.5 mm of powder sample thickness at a constant probesample distance of approximately 1 cm. These experiments
showed that the signal returning to the detector comes approximately from the top 0.5 mm of the powder sample. The
approximate sample volume when 12 scans are averaged is
312 mg and was calculated from the equation below which
takes into consideration that the sample is moving while the
spectra are being obtained.
"  
#
d 2
M
d x H
2

J Pharm Innov (2014) 9:291301

293

Fig. 1 Schematic of the


experimental setup for the
continuous mixing experiments: a
two pilot plant powder feeders, b
custom-made continuous mixer, c
conveyor belt, d NIR
spectrometer, and e sampling
point for reference method

where is the sample bulk density (0.60 g/cm3), d is the


NIR beam diameter, x is the sample displacement, and H is
the experimental depth of penetration of the NIR beam.

Preparation of Calibration and Validation Blends


A total of five calibration blends and three validation
blends were prepared spanning a concentration range
from 7 to 13 % w/w ibuprofen (Ibu). The Solver Excel
tool was used to create an experimental design that
reduced correlation between the concentrations of the
main excipients and ibuprofen.
All eight blends were individually prepared by placing
ibuprofen over a standard sieve (USA Standard Testing Sieves
ASTM Specification: Sieve Designation No. 30). Colloidal
silicon dioxide (SiO2) was placed over the ibuprofen along
with a portion of preweighed lactose in order to prevent the
cohesive mixture from sticking to the sieve. Components were
rubbed together through the sieve [13]. This preliminary mixture was used as the API preblend. A 16-qt PK Shell VBlender was charged with the components in the following
order: remaining preweighed lactose (Lac), API preblend, and
microcrystalline cellulose (MCC). The V-Blender was operated at 15 rpm for a total time of 60 min. Afterward, magnesium
stearate (MgSt) was added and the blend mixed for an additional 4 min. The API content in each sample was calculated
from the values obtained by weighing. The blending endpoint
of the calibration blends was monitored by calculating the
standard deviation of the 1,202-nm peak intensity at 12 different points of the blend after first derivative pretreatment for
every 15 min during the 60-min preblending process [7].

Preparation of Continuous Blending Process Preblends


Two preblends were individually prepared for the continuous
blending process: the API preblend and the Excipient
preblend. The API preblend was prepared as described above
and placed in the 16-qt PK Shell V-Blender operated at 15 rpm
for a total time of 30 min. The excipient preblend was prepared by charging the excipient components in the following
order: preweighed lactose (Lac), API preblend, and MCC.
The V-Blender was operated at 15 rpm for a total time of
60 min. Afterward, magnesium stearate (MgSt) was added
and the blend mixed for an additional 4 min.
NIR Calibration Model Development and Data Analysis
In-line NIR spectra of the calibration blends were obtained as
the powder moved underneath the NIR beam on the moving
conveyor belt as shown in Fig. 1. Data pretreatment and NIR
calibration model development was performed using SIMCAP (Umetrics Software Prediction Engine) and the partial least
square (PLS) algorithm. The NIR calibration model was developed after evaluation of spectral regions, data pretreatments, and number of PLS components. The calibration
models performance was first assessed in terms of root mean
standard error for calibration (RMSEC). The calibration
models performance was also evaluated in terms of root mean
standard error of prediction (RMSEP) using a prediction sample set (Eq. 3).
v
2
u Xm 
u
b
y
y
t i1 i i
RMSEC
2
N f 1

294

RMSEP

J Pharm Innov (2014) 9:291301

v
2
u Xm 
u
t i1 byi yi
N

where byi is the API concentration predicted by the NIR


calibration model, yi is the reference (gravimetric) concentration, N is the number of samples in the prediction or validation
sets, and f is the number of factors used in the model. All
spectra were mean centered. The spectral pretreatments tested
for model construction included first and second derivative
and standard normal variate (SNV). Derivatives were obtained
by applying the Savitzky-Golay (SG) algorithm using a 21point moving window [14, 15]. The effect of different spectral
regions was also evaluated.
Variograms were calculated using in-house developed software with MATLAB (The MathWorks, Natick, MA) version
7.10 (R2010a). Fast Fourier transform (FFT) analysis was
performed with a MATLAB discrete Fourier transforms
(DFT) to study the variability observed in the continuous
manufacturing results.

Fig. 2 Raw NIR spectra for the three major components in the pharmaceutical blend: ibuprofen, lactose, and microcrystalline cellulose. The
shaded area corresponds to the spectral region associated with the API
spectral bands

Results and Discussion


Ibuprofen UV Method
Development of NIR Calibration Model
To confirm the prediction of the NIR calibration model,
an in-house validated UV method was used as the
reference method for the quantification of the ibuprofen.
The absorbance of samples and standards was measured
at 266 nm with a 10-mm optical path length. Each
powder sample collected was weighed and transferred
to 500-mL volumetric flasks using a solution of 75 %
methanol: 25 % distilled water as diluent. After sample
preparation, the solution was analyzed using a Thermo
Electron Corp. UV spectrometer (Marietta, OH). A day
of use accuracy assessment was performed at the beginning of the analysis over three concentration levels to
evaluate the systems and the analysts performance.
Readings were made in duplicate for accuracy standards
and samples. A standard deviation of 0.9 % and an
average recovery of 100.7 % (n=36) for ibuprofen were
obtained. The methods linearity study showed a correlation coefficient (r2) of 1.000 over the range of 0.05
0.70 mg/mL ibuprofen.

NIR spectra for the main components of the formulation (Ibu,


Lac and MCC) were measured off-line as illustrated in Fig. 2.
Ibuprofen (API) is characterized by a group of absorption
bands at low wavelengths corresponding to the first and
second overtone vibrations, while the major excipients have
absorbance bands at higher wavelengths. The API concentration is the greatest source of variation in the 1,0901,428-nm
wavelength region as shown in Fig. 3. Well-defined clusters
can be seen in the PCA score plot for each API concentration

Blend Sampling
Samples were retrieved at the end of the conveyor belt
for each test sample set and continuous mixing run. The
flowing powder coming out at the end of the conveyor
belt was sampled every 5 s using a plastic cup to
sample the flowing stream obtaining approximately 3 g
of powder per time point [16]. Afterward, API quantification of each sample was performed off-line by the
in-house validated UV method.

Fig. 3 PCA score plot for the calibration sample set in the 1,0901,428nm wavelength region

J Pharm Innov (2014) 9:291301

295

Table 1 Parameters of the NIR calibration models constructed and evaluated


NIR calibration model no.

Spectral range
(nm)

Data pretreatment

Principal Components (#)

Percent of variation
R2Ycum

RMSEC
(%)

RMSEP
(%)

1
2
3
4

1,0901,605
1,0901,428

SNV+1st derv
1st derv
1st derv
SNV+1st derv

1
1
2
1

87.1
99.0
99.5
93.6

0.77
0.22
0.15
0.54

0.79
0.27
0.24
0.55

SNV+1st derv
1st derv
1st derv
SNV+1st derv
SNV+1st derv

2
1
2
1
2

97.9
95.0
98.9
94.6
98.5

0.31
0.48
0.22
0.50
0.26

0.37
0.59
0.40
0.59
0.42

5
6
7
8
9

1,0901,255

NIR near infrared, RMSEC root mean standard error for calibration, RMSEP root mean standard error of prediction, derv derivative, SNV standard normal
variate

level when using first derivative pretreatment, capturing


93.8 % of the spectral variation between the first two factors.
Random sample selection was used to select 40 % of the
calibration sample set spectra to develop the NIR calibration
model and the remaining 60 % as the prediction sample set.
Calibration models developed with several spectral regions
and number of PLS factors were evaluated to optimize the
ability of the NIR calibration model to predict the prediction
sample set. This first assessment was based on the RMSEC
(Eq. 2) and the RMSEP (Eq. 3). Table 1 shows the fraction of
the variation of the Y variables explained by the model
(R2Ycum) and the error of prediction for the prediction sample
set for some of the NIR calibration models evaluated. Not all
NIR calibration models were included for clarity purposes.
Results showed that when a wide spectral region (model
no. 1) was used to develop the NIR calibration model, the first
PLS factor describes less variation of the Y variable (API
concentration) than models developed using shorter spectral
range (model nos. 2, 4, 6, and 8). This is likely related with the

inclusion of high wavelengths into the model, which are


associated with the excipients of the formulation. The calibration model with 2 PLS factors provided a better prediction of
the Y variable as seen by the R2Ycum. Therefore, when comparing shorter spectral regions, 1,0901,428-nm spectral region and 2 PLS factors (model nos. 3 and 5) are determined as
the optimal parameters to develop the preliminary NIR calibration model with an average RMSEP of 0.3 % (w/w) as
shown in Table 1.
Method Robustness
The amount of blend over the conveyor may change over time
in the continuous manufacturing setup used in this process
(Fig. 1). The distance between probe and sample will vary as a
consequence of powder flow rate changes in the process. The
probe distance to the powder will be reduced when more
powder is accumulated or increased if less powder is present
in the conveyor belt. The effect of this variation was assessed

Table 2 Probe to sample distance effects in NIR model predicted concentration


Sample-probe distance (cm)

NIR calibration model response


Model no. 3 (1st derivative)

0.79
0.83
0.90
0.97
1.10
1.19
1.29

Model no. 5 (SNV/1st derivative)

Average predicted concentration % (w/w)

SD (n=6)

Average predicted concentration % (w/w)

SD (n=6)

8.90
9.04
9.24
9.51
9.89
10.20
10.56

0.04
0.05
0.09
0.08
0.09
0.12
0.04

8.29
8.27
8.41
8.48
8.40
8.38
8.48

0.16
0.17
0.07
0.08
0.16
0.16
0.14

NIR near infrared, SNV standard normal variate

296

J Pharm Innov (2014) 9:291301

by the NIR calibration model. The additional error did not


affect the NIR predictions.

Prediction of Validation Blends

Fig. 4 Comparison of NIR calibration model response when using SNV


pretreatment

by varying the probe to sample distance to a maximum of


0.5 cm and determining its effect on the predictions. Table 2
presents the NIR predicted concentrations of API and repeatability assessment for both preliminary NIR calibration
models at different probe to sample distance.
The results show that using first derivative to pretreat the
data lead to concentration predictions varying up to 1.66 % w/
w when the probe to sample distance varied 0.5 cm. On the
contrary, when using SNV prior to first derivative, the concentration predictions varied as little as 0.2 % w/w as the probe
to sample distances varied. Figure 4 illustrates the drug concentration predictions as a function of probe to sample distance. The predictions remained practically invariant when
SNV-first derivative is performed. All remaining predictions
were performed with SNV-first derivative because of the
robustness of this NIR calibration method to variations in
powder level and powder distance to NIR probe.
The effect of instrument noise on PLS predictions was also
evaluated as part of method robustness. The background noise
spectra acquired from the polymer standard were multiplied
by a factor of 3 and added to the spectra obtained from a 10 %
(w/w) API target blend. These new spectra were then predicted

The NIR calibration model was further evaluated for precision


and accuracy. Three independent validation blends of known
concentrations ranging from 9 to 11 % (w/w) API were predicted as they passed through the continuous manufacturing
setup. For each run, the feeder was filled with a validation
blend. The blend was passed through the blender, and NIR
spectra were obtained for approximately 4 min as the powder
moved over the conveyor belt. Blend samples were obtained
every 5 s for the determination of API by UV analysis [17].
Table 3 shows the excellent results obtained. Figure 5 further
illustrates the NIR predictions obtained from the validation
blends and the UV results measured at certain time points.
The largest difference between UV and NIR results was
0.36 % (w/w), for the 11 % (w/w) blend. Close evaluation of
these experimental runs showed that after steady-state condition (approx. 80 to 90 s), average NIR predictions from the 9
and 10 % (w/w) validation blends were within the 95 %
conflict of interest (CI) of the average target concentration
range. The average NIR prediction of the 11 % validation
blend is within the 99 % CI (0.36) of the average target
concentration range. Therefore, results between the two
methods were comparable.
The validation blends also provided valuable information
in terms of method precision. The standard deviations obtained by the NIR method were NMT 0.2 % and with an RSEP
below 4 %. The standard deviation for the validation blends is
two to three times larger than those observed for the repeatability studies shown in Table 3. The repeatability studies
were performed by obtaining six consecutive spectra without
moving the sample (same sample) and ranged from 0.08 to
0.17 % (w/w) (Table 2). Thus, the repeatability study
represents the method error, the minimum error that
may be expected for the NIR method. Since the blends
were well mixed before they were placed in the feeder,
the 0.2 % (w/w) standard deviation is considered the
minimum variation that could be expected in the

Table 3 NIR model prediction of validation blends


API target concentration % (w/w) NIR

UV

Predicted API mean % (w/w) SD


9
10
11

8.89
9.61
11.17

RSEP (%) CI (95 %) Measured API mean % (w/w) SD

0.24 2.81
0.20 2.82
0.21 3.93

0.02
0.02
0.02

NIR near infrared, API active pharmaceutical ingredient, RSEP relative standard error of prediction

8.78
9.81
10.81

CI (95 %)

0.47 0.27
0.43 0.25
0.49 0.28

J Pharm Innov (2014) 9:291301

297

Fig. 5 NIR predictions obtained from the monitoring of three validation blends. UV measurements of API content for each run are also illustrated

continuous manufacturing setup discussed in this study.


However, the 0.2 % (w/w) standard deviation was obtained when the blends were preblended, passed through
one feeder, through the mixer, and deposited over the
conveyor belt. The next phase of the study included the
entire system where the two feeders were used.

expected. These are considered excellent preliminary results


since the choice and operation of the custom-made mixing
equipment still require optimization [5].
Table 4 also reveals that NIR predicts a higher drug concentration than that determined by the UV method. This
difference was not observed in the validation blends (Table 3)

Real-time Monitoring of Continuous Blending Process


The validated calibration model was used to monitor a continuous blending process for 3 min. Figure 6 shows the realtime monitoring of a continuous powder blending operation.
The system was set up to target a 10 % w/w ibuprofen blend.
Blend samples were collected for the corresponding off-line
UV analysis. The NIR method predicted the API concentration approximately every 0.24 s. The UV samples were collected approximately every 60 s, and the results obtained are
shown in Table 4.
All individual predictions were within 3 standard deviations of the average after steady-state conditions were
achieved. The standard deviation for the NIR predictions
was 0.49 % w/w. The standard deviation was approximately
two times greater than obtained for the validation blends.
Since the validation blends were premixed before being monitored in the continuous manufacturing setup, the increase in
the standard deviation in the continuous blending process was

Fig. 6 NIR predictions obtained from real-time monitoring of a continuous blending process. UV measurements of API content are also
illustrated

298

J Pharm Innov (2014) 9:291301

Table 4 Evaluation of the continuous blending process


NIR

UV

Predicted API mean % (w/w)

SD

CI (95 %)

Measured API mean % (w/w)

SD

CI (95 %)

10.5

0.49

0.04

9.3

0.71

0.23

NIR near infrared, API active pharmaceutical ingredient

and is not understood at the present time. The main difference


in the experimental setup is that the full continuous
manufacturing system used two feeders (for API and excipient), while the setup for the validation blends used only one
feeder. These results are in the same range of prediction
concordance as the results obtained by monitoring real-time
on-line blend uniformity by Sulub et al., which obtained an
average prediction difference of 2.2 % between API NIR
predictions (%) and HPLC (%) measurements [18]. This is
further evidence of the difficulty of analyzing blends. A
careful review of previous publications revealed that very
few studies have compared real-time NIR measurements and

off-line measurements [6, 1921]. This comparison is not easy


since the NIR method is providing the concentration of a
sample volume that is approximately 300 mg while the sample
analyzed by UV is about 3 g. The authors recognize that
additional experimental work is required to evaluate the difference between UV and NIR results.
The variation after steady state was also evaluated with the
use of a variogram. The theory of sampling defines onedimensional (1-D) heterogeneity for processes where samples
are obtained along one dimension in space, and there is a
natural linear order among the increments [22]. In continuous
manufacturing, the drug concentration in the blend may vary

Fig. 7 Variographic analysis of the validation blends and the real-time monitoring of the continuous blending process

J Pharm Innov (2014) 9:291301

299

over time, and time constitutes the dominant dimension. The


variogram was used to optimize the NIR sampling rate for the
process. The variogram was used to identify trends in the
process data by calculating the correlation between NIR predictions [23]. Thus, a lag of 1 measures the correlation between consecutive NIR predictions, and a lag of 2 measures
the correlation between every other NIR prediction.
V j

N
2
1 X 
aN j aN
2N j N1

where N is the number of data points in the time series (or


lot), j is the lag (which refers to the distance in time between
two NIR predictions, and a is the concentration. Low values of
V(j) result from similar drug concentration predictions and
indicate high correlation between the NIR predictions.
Figure 7 shows the variograms obtained for the NIR predictions from the continuous blending process and the validation blends. The lag (inter-distance) between any two measurements is measured in seconds (equidistant intervals). The
variogram function, V(j), was evaluated from 1 to 100 increments. Each variogram shows a cyclic behavior as well as a
slight increase in the variogram function (V(j)) as the lag
becomes increasingly larger. A cyclic variogram is typically
observed when the variability of the process is influenced by a
factor causing this behavior. This is representative of the
quasi-steady state of the system and demonstrates that the

variogram analysis is an effective tool to determine if the


system is operating in steady state.
In the validation blend runs, results for V(j) and the change
in the autocorrelation function are minimal (approx. 0.04) for
consecutive lag times since these blends are less heterogeneous (previously blended) when compared to the continuous
blending run. In the validation runs, only one feeder was used,
and the ascending tendencies in the variograms were related to
small changes in powder feed rate affecting the distance
between the NIR sensor and the powder bed.
During the continuous blending run, the autocorrelation
function varied approximately ten times (approx. 0.4) more
than for the validation blends describing more heterogeneity
between consecutive measurements. In addition, the continuous blending run variogram increases less rapidly with increasing lag time compared to the validation blend runs. The
autocorrelation function varies from 0.4 and 0.9, but with
minimal change from a lag time of 10 to values as high as
100. This low variation may be a result of steady state. During
the continuous blending run, two feeders were used; therefore,
more powder remained in the feeder extending the time to get
to the emptying stage, but additional studies are necessary to
confirm this observation. This run also shows a minimum at
lag (j)=21 and additional minima at j=35, 48, 61, 74, 84. This
indicates a period of 13 NIR measurements (approximately
3.1 s). According to the Nyquist theorem, it is recommended
to sample at a higher frequency than two per period to capture
the process variation. NIR measurements of the continuous

Fig. 8 FFT analysis of the validation blends and the real-time monitoring of the continuous blending process

300

process could be taken every 1.4 s, more than half the observed frequency of 3.1 s. In future experiments, the sampling
rate will be set to the optimized parameters found. Thus, the
variogram is a useful tool for determining how frequently to
sample.
FFT analysis was performed to study the variability observed in the continuous manufacturing results. Figure 8
shows the frequencies associated to the measurements of
NIR API prediction concentration obtained through time. In
this continuous manufacturing setup, the screw feeders [24]
and the mixer influence in the observed cyclic behavior. Two
dominant frequencies at approximately 0.98 and 1.32 Hz were
observed in the validation runs. These frequencies could be
associated to the mixer and screw feeder speed cyclic behavior, respectively. These units were operated under the same
conditions in all validation runs. Three dominating frequencies were observed in the continuous blending run. One of
dominating frequencies (0.98 Hz) was also observed in the
validation runs. This observation is not unexpected since the
mixer conditions were the same as for those runs. The other
two dominating peaks (0.62 and 0.96 Hz) were unique for the
continuous run. This could probably be associated to the use
of two feeders in the continuous blending process. Moreover,
both peaks are at lower frequencies compared to the 1.32 Hz
associated with the screw feeder speed in the validation runs.
This is in agreement with the fact that during the continuous
blending process, the screw feeder speed in feeder #2 was set
at a lower RPM to maintain the total feed rate since an
additional feeder (feeder #1) was used. Therefore, the two
0.62 and 0.96 Hz peaks can be associated to the two different
screw feeder speeds used in the continuous blending run.
Thus, the use of FTT complemented the variogram results in
identifying the root causes of the cyclic behavior observed.

Conclusions
The sources of error in the NIR method used to monitor a
continuous mixing process have been investigated. Repeatability studies measured the instrumental error that ranged
from 0.07 to 0.17 % (w/w). The precision and accuracy in
determining well-mixed blends were also determined by monitoring previously mixed blends as they passed through the
continuous manufacturing setup. The standard deviation of
the well-mixed blends was about 2.5 times greater than the
variation observed in the repeatability study. This increase in
standard deviation is due to variations in the composition of
the powder mixture and powder flow. Powder flow differences affect the level of powder over the conveyor belt and
vary the distance between the powder and the NIR instrument.
NIR predictions were affected by the probe to powder distance
in the initial calibration models. However, the use of the SNV

J Pharm Innov (2014) 9:291301

transform significantly reduced the effect of differences in


powder flow on the NIR predictions. The standard deviation
of the continuous mixing experiment ranged from 0.47 to
0.53 % (w/w) about twice the variation observed for the
validation mixtures. The increase is associated with the use
of two feeders and the mixing of excipients and API. This
thorough characterization of the precision of the NIR method
is necessary in continuous manufacturing where the NIR
system is used to monitor drug concentration after steady state
is achieved.
The cyclic variation observed in the variograms and the
results from the analysis of the FFT can identify the units in an
operation causing variation in the measurements in this continuous manufacturing setup. The variogram and the FTT
showed to be effective tools to investigate the variability of
the blend under study and present an approach to understand
how frequently a continuous process has to be sampled.
Acknowledgments This study was financially supported by the NSF
Engineering Research Center for Structured Organic Particulate Systems
(EEC-0540855). The authors would like to thank Jesus Torres for assistance with the Fast Fourier Transform analysis that greatly improved the
manuscript.

References
1. Bell TA. Challenges in the scale-up of particulate processesan
industrial perspective. Powder Technol. 2005;150(2):6071.
2. Leuenberger H. New trends in the production of pharmaceutical
granules: the classical batch concept and the problem of scale-up.
Eur J Pharm Biopharm. 2001;52(3):27988.
3. Mort PR. Scale-up of binder agglomeration processes. Powder
Technol. 2005;150(2):86103.
4. Rodriguez-Justo O, Morales M. Analysis of process parameters on
the characteristics of liposomes prepared by ethanol injection with a
view to process scale-up: effect of temperature and batch volume.
Chem Eng Res Des. 2011;89(6):78592.
5. Florian M, Velzquez C, Mndez R. New continuous tumble mixer
characterization. Powder Technol. 2014;256:18895.
6. Jamrogiewicz M et al. Determination of API content in a pilot-scale
blending by near-infrared spectroscopy as a first step method to
process line implementation. Acta Pol Pharm. 2013;70(3):41929.
7. Shi Z et al. Process characterization of powder blending by nearinfrared spectroscopy: blend end-points and beyond. J Pharm
Biomed Anal. 2008;47(45):73845.
8. Liew CV, Karande AD, Heng PW. In-line quantification of drug and
excipients in cohesive powder blends by near infrared spectroscopy.
Int J Pharm. 2010;386(12):13848.
9. Sekulic SS et al. On-line monitoring of powder blend homogeneity
by near-infrared spectroscopy. Anal Chem. 1996;68(3):50913.
10. Momose W et al. Process analytical technology applied for end-point
detection of pharmaceutical blending by combining two calibrationfree methods: simultaneously monitoring specific near-infrared peak
intensity and moving block standard deviation. Powder Technol.
2011;210(2):12231.
11. Vanarase AU et al. Real-time monitoring of drug concentration in a
continuous powder mixing process using NIR spectroscopy. Chem
Eng Sci. 2010;65(21):572833.

J Pharm Innov (2014) 9:291301


12. Martnez L et al. Use of near-infrared spectroscopy to quantify drug
content on a continuous blending process: influence of mass flow and
rotation speed variations. Eur J Pharm Biopharm. 2013;84(3):606
15.
13. Beach L et al. Near-infrared spectroscopy for the in-line characterization of powder voiding part II: quantification of enhanced flow
properties of surface modified active pharmaceutical ingredients. J
Pharm Innov. 2010;5(12):113.
14. Barnes RJ, Dhanoa MS, Lister SJ. Standard normal variate transformation and de-trending of near-infrared diffuse reflectance spectra.
Appl Spectrosc. 1989;43(5):7727.
15. Rinnan , Berg Fvd, Engelsen SB. Review of the most common preprocessing techniques for near-infrared spectra. TrAC Trends Anal
Chem. 2009;28(10).
16. Popo M et al. Blend uniformity analysis using stream sampling and
near infrared spectroscopy. AAPS PharmSciTech. 2002;3(3):E24.
17. Andersson M et al. NIR spectroscopy on moving solids using a
scanning grating spectrometerimpact on multivariate process analysis. Chemom Intell Lab Syst. 2005;75(1):111.

301
18. Sulub Y et al. Real-time on-line blend uniformity monitoring using
near-infrared reflectance spectrometry: a noninvasive off-line calibration approach. J Pharm Biomed Anal. 2009;49(1):4854.
19. Koller DM et al. Continuous quantitative monitoring of powder
mixing dynamics by near-infrared spectroscopy. Powder Technol.
2011;205(13):8796.
20. El-Hagrasy AS et al. Near-infrared spectroscopy and imaging for the
monitoring of powder blend homogeneity. J Pharm Sci. 2001;90(9):
1298307.
21. Berthiaux H, Marikh K, Gatumel C. Continuous mixing of powder
mixtures with pharmaceutical process constraints. Chem Eng Process
Process Intensif. 2008;47(12):231522.
22. Bakeev KA. Process analytical technology: spectroscopic tools and
implementation strategies for the chemical and pharmaceutical industries. Wiley; 2005.
23. Petersen L, Esbensen KH. Representative process sampling for reliable data analysisa tutorial. J Chemom. 2005;19(1112):62547.
24. Engisch WE, Muzzio FJ. Method for characterization of loss-inweight feeder equipment. Powder Technol. 2012;228:395403.

Das könnte Ihnen auch gefallen