You are on page 1of 14

Journal of Pharmaceutical Sciences 105 (2016) 168e181

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


journal homepage: www.jpharmsci.org

Pharmaceutics, Drug Delivery and Pharmaceutical Technology

Integrated Application of Quality-by-Design Principles to Drug


Product Development: A Case Study of Brivanib Alaninate
FilmeCoated Tablets
Sherif I.F. Badawy 1, *, Ajit S. Narang 1, Keirnan R. LaMarche 1,
Ganeshkumar A. Subramanian 1, Sailesh A. Varia 1, Judy Lin 2, Tim Stevens 2,
Pankaj A. Shah 2
1
Drug Product Science and Technology, Bristol-Myers Squibb Company, New Brunswick, New Jersey 08903
2
Analytical and Bioanalytical Development, Bristol-Myers Squibb Company, New Brunswick, New Jersey 08903

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

Article history: Modern drug product development is expected to follow quality-by-design (QbD) paradigm. At the same
Received 2 July 2015 time, although there are several issue-specific examples in the literature that demonstrate the appli-
Revised 8 October 2015 cation of QbD principles, a holistic demonstration of the application of QbD principles to drug product
Accepted 10 November 2015
development and control strategy, is lacking. This article provides an integrated case study on the sys-
tematic application of QbD to product development and demonstrates the implementation of QbD
concepts in the different aspects of product and process design for brivanib alaninate filmecoated
Keywords:
tablets. Using a risk-based approach, the strategy for development entailed identification of product
quality by design
tablets
critical quality attributes (CQAs), assessment of risks to the CQAs, and performing experiments to un-
brivanib alaninate derstand and mitigate identified risks. Quality risk assessments and design of experiments were per-
design space formed to understand the quality of the input raw materials required for a robust formulation and the
control strategy impact of manufacturing process parameters on CQAs. In addition to the material property and process
critical quality attributes parameter controls, the proposed control strategy includes use of process analytical technology and
critical process parameters conventional analytical tests to control in-process material attributes and ensure quality of the final
critical material attributes product.
dissolution
© 2016 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
porosity

Introduction established acceptance criteria for the critical quality attributes


(CQAs).1,2 Although QbD principles were introduced more than a
The principles of Quality by Design (QbD) emphasize the need decade ago by the ICH guidelines, there are hardly any published
for understanding sources of variability in the manufacturing pro- examples demonstrating how these principles are reduced to
cess and how it links to product quality as prerequisite for practice in the commercial development of a drug product as well
robustness. Risk-based approach is used to define the goals of QbD as transfer and implementation of control strategy in a
development and to design appropriate development studies to manufacturing plant. Although there are some published reports
achieve these goals. Product quality attributes based on patient invoking QbD principles in the study of a specific issue,3-11 none of
needs and clinical relevance are identified, and the risks to these them provide a wide view of the application of QbD to a compre-
attributes due to input material variability and manufacturing hensive development program.
process parameters are assessed. The output of QbD development Brivanib alaninate (BMS-582664) is a potent, orally active
takes the form of a process and product quality control strategy inhibitor of vascular endothelial growth factor receptors.12 A film-
which ensures that the final product consistently meets pre- coated tablet formulation for brivanib alaninate has been devel-
oped using a wet granulation manufacturing process. The
principles of QbD were applied during formulation and process
* Correspondence to: Sherif I. F. Badawy (Telephone: þ1-732-227-6451; Fax: 732-
227-5150).
development of brivanib alaninate filmecoated tablets. Using a
E-mail address: sherif.badawy@bms.com (S.I.F. Badawy). risk-based approach, the strategy for the formulation design and

http://dx.doi.org/10.1016/j.xphs.2015.11.023
0022-3549/© 2016 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 169

understanding was developed by (1) identifying desired product Table 1


attributes, (2) identifying quality risks to the attributes during Experimental Design for the Formulation Optimization Study

development, and (3) adopting the appropriate strategy and per- Batch Croscarmellose Hydroxypropyl Crospovidone Magnesium
forming experiments to mitigate or reduce risks. Quality risk as- Number Sodium (%) Cellulose (%) (%) Stearate (%)
sessments and statistically designed experiments were performed 1 3.0 3.0 3.0 1.0
to understand the quality of the input raw materials required for a 2 3.0 3.0 3.0 0.5
robust formulation and the impact of manufacturing process pa- 3 4.5 1.5 1.5 1.5
4 4.5 1.5 1.5 0.5
rameters on the CQAs of the drug product. The data obtained were
5 1.5 1.5 4.5 1.5
used to establish acceptable material attribute and process ranges 6 1.5 1.5 4.5 0.5
and to define the control strategy for the commercial manufacture 7 4.5 4.5 4.5 1.5
of brivanib alaninate filmecoated tablets. The proposed commer- 8 4.5 4.5 4.5 0.5
9 4.5 1.5 4.5 1.5
cial manufacturing process uses process analytical technology and
10 4.5 1.5 4.5 0.5
conventional analytical tests to control the in-process material at- 11 1.5 4.5 1.5 1.5
tributes. The product quality is controlled and assured through a 12 1.5 4.5 1.5 0.5
holistic approach of input material, in-process attribute, and 13 1.5 4.5 4.5 1.5
parametric controls combined with appropriate finished product 14 1.5 4.5 4.5 0.5
15 4.5 4.5 1.5 1.5
tests. The purpose of this article is to provide an integrated case
16 4.5 4.5 1.5 0.5
study on the systematic application of QbD to product development 17 3.0 3.0 3.0 1.0
and to demonstrate the implementation of QbD concepts in the 18 3.0 3.0 3.0 1.0
different aspects of product and process design. The work pre- 19 1.5 1.5 1.5 1.5
sented demonstrates how QbD concepts, such as product robust- 20 1.5 1.5 1.5 0.5

ness and scale-independent design space, were reduced to practice


in the development of brivanib alaninate tablets.
DoE batches were manufactured in a Fuji high shear granulator
Methods with a 25-L bowl at 5 kg total batch size. The impeller speed was
maintained at 240 rpm (equivalent tip speed of 4.8 m/s), chopper
Formulation Selection run at low speed (1000 rpm). A peristaltic pump was used to add
water at a rate of 100 g/min/kg over a period of 4 min and 30 s. The
Formulation and process screening studies were conducted to wet massing time was kept constant at 15 s for all batches while
select a prototype formulation and manufacturing process for bri- maintaining same impeller and chopper speeds. The wet granula-
vanib alaninate tablets. Key formulation selection studies focused tion was passed through a conical mill (Comil 197S) with 0.125-inch
on the selection of tablet disintegrant in which performance of both (3.2 mm) round opening screen at impeller speed of ~1350 rpm.
croscarmellose sodium (CCS) and crospovidone (CPVP) was evalu- The granulation was then dried using the GPCG-1 fluid-bed dryer at
ated. Formulation selection batches were manufactured at 600 g 70 C to a moisture content of 1.50%. The dried granulation was
batch size by a wet granulation process in a 4-L high-shear Diosna milled using a conical mill (Comil 197S) with 0.045-inch (1.1 mm)
granulator (Diosna Dierks & So €hne GmbH, Osnabrück, Germany). screen opening at impeller speed of ~1350 rpm. The milled gran-
All batches were granulated using 45%-50% w/w water, relative to ulation was split into 2 equal sublots and then placed in a bin
total batch size, at impeller speed of 5.2 m/s. Wet granulation was blender (8.3 L) and mixed with the extragranular excipients and
screened through 8 mesh screen, and granules were subsequently appropriate amount of magnesium stearate per the experimental
dried in a Glatt GPCG-1 fluid-bed dryer (Glatt Air Techniques, design. The final blend was compressed into 400 mg strength
Ramsey, NJ) until the loss on drying (LOD) was 2% w/w. Dried tablets (800 mg tablet weight) on the 6 station Korsch PH 100 tablet
granulations were milled using a conical mill (Comil 197S, Quadro, press using embossed tooling. Batches were characterized using
Waterloo, Ontario, Canada) and then blended with the extra- methods described below. Statistical analysis of response data was
granular excipients in a bin blender. The final blend was com- performed by regression analysis in SAS JMP (SAS Institute Inc.,
pressed into 400 mg of strength tablets (800 mg tablet weight) on a Cary, NC).
6 station Korsch PH 100 tablet press (Korsch AG, Berlin, Germany)
using embossed tooling. Batches were characterized using methods QbD Methodology
described below.
After formulation composition was established, studies were
Formulation Optimization conducted to evaluate effect of material attributes and process
parameters on product attributes of the selected formulation. CQAs
After the prototype formulation was identified, a formulation for brivanib alaninate tablets were identified as tablet potency,
design of experiment (DoE) study was conducted to assess dosage uniformity, impurities, dissolution rate, and appearance.
formulation ruggedness and optimize composition with respect to Appearance was considered a CQA as it is perceived to affect patient
the binder (hydroxypropyl cellulose [HPC]), disintegrant (intra- and compliance and hence outcome of therapy.
extragranular), and lubricant (magnesium stearate). A split-plot Subsequent to the identification of the CQAs, failure mode effect
full-factorial experimental design was used to complete the DoE. analysis was conducted to identify material attributes and process
The design entails the manufacture of a larger scale granulation parameters to be studied for their impact on CQAs. Although all
batch and splitting the batch into 2 parts to study the 2 levels of material attributes and process parameters that can potentially
magnesium stearate, instead of manufacturing separate batches for affect CQAs were considered as part of the risk analysis, only a
each combination. This design reduces the number of granulation subset of these attributes and parameters were selected for
batches that need to be manufactured by half, while still providing development studies as warranted by the risk analysis. Studies
sufficient information to understand the main effects and in- were conducted to generate knowledge regarding the effect of the
teractions of the various excipients. Experimental design batches selected input material attributes and process parameters on CQAs
are shown in Table 1. and identify critical material attributes (CMAs) and critical process
170 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

data were used to determine compactibility parameter (initial slope


of the compaction profile), hardness to force ratio at target tablet
hardness, and maximum hardness achieved for each batch.

Granule Porosity
Pore distribution of granulation and tablets was determined by
mercury intrusion porosimetry (MIP, AutoPore III; Micromeritics,
Norcross, GA). A fraction of the tablet or a granulation sample of the
particle size fraction passing through 60-mesh screen and retained
on 100-mesh screen was used for the porosity determination.
Incremental pore volume was determined at different pressures
ranging from 1 to 61,000 psi. The pore diameter corresponding to a
given pressure was calculated by the software of the instrument
according to the Washburn equation using mercury surface tension
value of 485 dyne/cm and mercury contact angle of 130 . Pores in
the 0.1- to 10-mm range were used for the assessment and com-
parison of the different batches.

Tablet Appearance
Figure 1. QbD methodology applied to the development of brivanib alaninate tablets. Compression runs for all batches were monitored for sticking to
the tablet compression tooling and defects on the tablets. Tooling
was removed after each run to assess for sticking, filming, and
parameters (CPPs) based on these studies. Critical attributes and filling of the embossing. A scoring system was devised to help
parameters are defined as such if their variation within reasonable quantify the extent of sticking on punches, and extent and fre-
range results in a measurable impact on any of the CQAs. Proven quency of defects on the tablets. The criterion for tablet picking was
acceptable ranges (PARs) were established for the CMAs and CPPs based on the severity and frequency of picking. A score of “0”
based on these development studies. Controlling CMAs and CPPs indicated no more than minor picking on <30% of tablets, whereas a
within their PARs, together with in-process tests and controls of score of “5” indicated severe picking in >30% of tablets. A scoring
intermediate materials, constituted the control strategy for the system was also established to rank order the formulation for
brivanib alaninate tablet manufacturing process (Fig. 1). tendency to stick to the punches, wherein no sticking was assigned
All non-CPPs which were either not identified for evaluation as a score of “0” and severe sticking that obliterated punch face
per the risk analysis or found to be noncritical from development emboss was assigned a score of “5.”
studies were not included in the control strategy. These parameters
were set at a fixed value at batch record level, and no PARs were Analytical Methods
established for these parameters.
Tablet Dissolution
Physical Characterization Brivanib alaninate is a Biopharmaceutics Classification System
class II compound with pH-dependent solubility and high perme-
Particle Size Distribution ability.13 Dissolution test of brivanib alaninate tablets was con-
The particle size distribution of the final granulation was ducted in 1000 mL of 50-mM phosphate buffer, pH 6.8, containing
determined by mesh analysis using an Allen-Bradley Sonic Sifter 1.0% Triton X-100 at 37 C using USP apparatus II at 75 rpm.
(Allan-Bradley, Milwaukee, WI) equipped with a series of 6 screens The target dissolution limit was not less than 75% dissolved after
and a pan. Approximately 5 g of the sample was tested with a pulse 30 min, which was based on the dissolution profiles of formulations
setting of “5,” sift setting of “5,” and total sifting time of 5 min. The used in the different stages of clinical development.
geometric mean diameter of the granulation was calculated from
the granulation weight fraction retained on each screen. The Moisture Methods of Blends and Tablets
percent fines (<75 mm) was calculated by the summation of the An LOD method with testing temperature at 105 C was used for
percent material retained over the 325-mesh screen and dust blends and tablets to determine moisture at the end of fluid-bed
collected in the pan of the sonic sifter. drying and during tablet coating.
A USP volumetric KarleFischer titration method was used to
Flow Properties determine the water content in the final film-coated tablets. The
Flow properties of the lubricated blend were tested using an tablets were ground to fine powder before adding to the
Erweka GTB powder flow tester (Erweka GmbH, Heusenstamm, KarleFischer titration vessel.
Germany). The time required for 100 g of the blend to flow through An on-line near infrared (NIR) spectroscopic method was also
10-mm aperture was determined and converted to a flow rate developed as an in-process control to measure and control moisture
in g/s. in brivanib granulation during fluid-bed drying using a Brimrose
4030-EX explosion-proof NIR spectrometer (Brimrose Corporation,
Compaction Properties Sparks, MD). The spectrometer is attached to a window on the side of
Presster compaction simulator (Metropolitan Computing Cor- the dryer bowl and is used to measure granule moisture through the
poration, East Hanover, NJ) was used to generate the compaction window in real time during the drying process. A chemometric
profile data at 14-ms dwell time using oval-shaped B tooling and model was developed and calibrated to water content (as % w/w) in
800 mg tablet weight. Different compaction forces were used to the granulation using results from LOD measurements of calibration
compress the granulation, and tablet hardness obtained at each standards determined by a moisture balance. The on-line NIR
compression force was plotted against the corresponding measurements determine when the material has reached the
compaction force to obtain the compaction profile. The Presster desired moisture level and provide the end point for the drying step.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 171

Calibration standards were made up of granules equilibrated to


varying moisture levels. During method development, additional
samples were created to test the robustness of the method with
respect to granule density, granule size, excipient and drug sub-
stance concentration, and drug substance particle size.

HPLC Potency and Impurity Method


Tablet potency and impurity were determined by HPLC reverse
phase method. Tablets were dispersed to fine powder in the
diluent, a mixture of acetonitrile, water, and acetic acid. Filtered
sample solutions were analyzed by an isocratic HPLC method for
potency and a gradient method for impurities using Zorbax SB-AQ
column (Agilent Technologies, Santa Clara, CA) and mobile phase
consisting of ammonium trifluoroacetate buffer, acetonitrile,
methanol, and trifluoroacetic acid. The tablet potency was deter-
mined based on the UV response of the active peak from the sample
solution and that from an external standard solution.
HPLC potency and impurity method was validated according to
ICH guideline Q2(R1), Validation of Analytical Procedures. The
validation results demonstrated that the specificity, linearity, ac-
curacy, precision, and robustness of the method were appropriate
for its intended use.

NIR Tablet Potency Method


The at-line NIR method was developed and validated in the
reflectance mode using a Thermo Antaris NIR spectrometer
(Thermo Fisher Scientific, Waltham, MA) in conjunction with a
chemometric model calibrated to brivanib alaninate concentration
(% w/w). Using prepared calibration tablet standards, the NIR
measurements were calibrated with results from the primary HPLC
tablet potency method computed from the average of 2 prepara-
tions of 5 tablets each. An appropriate partial least squares che-
Figure 2. Flow diagram of the manufacturing process for brivanib alaninate filme
mometric model was selected to ensure that the spectral variation
coated tablet.
was covariant with the brivanib concentration. The NIR method
computes % concentration of brivanib alaninate from 30 individual
compression tooling. Previously reported study showed that wet
tablet measurements, which is then adjusted to % label claim using
granulation reduces sticking of the blend to tablet compression
the individual tablet weights. The batch potency is computed from
tooling compared with a roller compaction process.14 The lower
the average of the 30 tablet results. Because the NIR method
sticking tendency of wet granulation over roller compaction was
measures only the surface of the tablet, a statistical analysis was
attributed to the differences in the ability of the granulation process
conducted to determine the appropriate number of tablets that
to modify exposed particle surface through the agglomeration of
would provide a similar assurance of batch potency as measuring
particles. The presence of greater proportion of primary drug par-
potency of 2 sets of 5 tablets by HPLC analysis.
ticles in the formulation is expected to exacerbate sticking, whereas
Tablet standards were prepared to build the calibration model for
the presence of agglomerated drug particles reduces sticking. Fines
the NIR method. The composition of the tablets was varied using a
in the wet granulated formulation are expected to be agglomerates
structured design so that obtained spectra would encompass ex-
of primary drug particles, in contrast to fines in roller-compacted
pected variations that may be encountered during routine use. The
samples which are largely unagglomerated primary drug particles.14
calibration standards were developed using a development granu-
lation batch and then through DoEs, extragranular excipient levels
were uniformly varied to achieve a drug substance calibration in-
dependent of extragranular excipient level. All calibration tablets
were manufactured using pilot-scale processes that mimicked the
commercial manufacturing process. Robustness was further built
into the model by identifying sources of method variation and either
testing these variations directly or incorporating them into the
calibration set of prepared tablets. Sources of method variation that
were identified and assessed included drug substance particle size,
granule size, tablet moisture content, tablet hardness, laboratory
humidity, and instrument-to-instrument variability.

Results

Formulation Selection and Optimization

Wet granulation process (Fig. 2) was selected to reduce the risk of Figure 3. Impeller current profile during granulation of formulations with different
drug substance adhesion to manufacturing equipment and disintegrants.
172 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

Table 2 The formulation of DoE study showed that higher level of HPC
Composition of Brivanib Alaninate FilmeCoated Tablets improves the formulation and process robustness by reducing the
Ingredient Function Prototype Final amount of fines in the granulation, which lowers its sticking
Formulation Formulation potential to tablet tooling and picking of tablets in compression. In
Concentration (% w/w) addition, higher amount of the lubricant (magnesium stearate)
reduced the risk of sticking and did not affect the dissolution rate.
Brivanib alaninate Active 50.00 50.00
(BMS-582664-02) The negative effect of magnesium stearate on compaction proper-
Microcrystalline Filler/binder 39.50 38.25 ties was minimal and of little practical significance. In addition, a
cellulose (PH102) 2-way interaction was observed between HPC and magnesium
Croscarmellose sodium Disintegrant 3.00 3.00
stearate (Fig. 4), which showed that formulations with lower binder
Hydroxypropyl cellulose Binder 3.00 4.00
Crospovidone Disintegrant 3.00 3.00 content have higher lubricant requirement. At the higher HPC level,
Colloidal silicon dioxide Glidant/compaction aid 0.50 0.50 the powder is better granulated resulting in magnesium stearate
Magnesium stearate Lubricant 1.00 1.25 being less critical to reducing sticking. However, at the lower level
Total, core tableta 100.0 100.0 of HPC, the powder is less granulated with more fines, and the
Opadry® (weight gain) Film coat 3.00 3.00
amount of magnesium stearate is critical to reduce sticking of
a
Total core tablet weight ¼ 800 mg. powder to the punch surface, hence resulting in the 2-way inter-
action between HPC and magnesium stearate. The outcome of the
study indicated that the formulation needs to be well-granulated to
Figure 3 shows impeller power consumption versus amount of minimize sticking risk. Accordingly, concentrations of HPC and
water added for formulations with CCS or CPVP as the intragranular magnesium stearate were increased in the optimized formulation
disintegrant. As water is added, impeller power increases as granule selected for the commercial product (Table 2).
growth takes place. A rapid increase in impeller power around
granulation end point is generally undesirable as it indicates rapid
CQAs, Process Parameters, and Material Attributes
uncontrolled granule growth which makes the process more diffi-
cult to control. The use of CCS as the intragranular disintegrant
Details for each CQA are presented below with respect to ma-
showed steady controlled granule growth, which increases the
terial attributes and process parameters assessed in development
robustness of the manufacturing process. In contrast, the use of CPVP
studies and those identified as critical based on development
as the intragranular disintegrant showed rapid granule growth and
studies. Fishbone diagrams, 1 for each CQA, are depicted below
“sharp” end point, which increases sensitivity to changes in process
showing the different parameters and attributes evaluated for their
parameters. This was shown by impeller power consumption mea-
impact on the CQA. Those identified as critical based on develop-
surement (Fig. 3) and visual observations during granulation.
ment studies are colored red, whereas noncritical attributes and
On the other hand, the use of CCS as a disintegrant was associ-
parameters are colored green.
ated with incomplete in vitro drug release. A previous study
showed that this was due to pH-dependent and reversible ionic
binding of brivanib alaninate by CCS. A pharmacokinetic study in In Vitro Drug Release (Tablet Dissolution)
monkeys indicated that bioavailability of brivanib alaninate is not Process parameters of individual unit operations and material
negatively impacted by presence of CCS.15 This in vitro binding attributes studied based on risk assessment of dissolution are
phenomenon was not observed with the use of CPVP as a dis- shown in Figure 5.
integrant. Although binding of brivanib alaninate by CCS was not
expected to be biorelevant based on this study, bioavailability risk Material Attributes and Dissolution. As shown in Figure 5, drug
posed by CCS still remained. Based on the findings from the wet substance particle size was evaluated for its effect on the dissolu-
granulation and CCS binding studies, CCS was selected as an tion rate for this BCS II compound. Tablet batches were manufac-
intragranular disintegrant to enhance robustness of the wet gran- tured at target process parameters using drug substance lots with
ulation process. However, CPVP was selected as the extragranular different particle size and evaluated for their dissolution behavior.
disintegrant to reduce total amount of CCS in the tablet and mini- Tablet dissolution rate was found to be dependent on drug sub-
mize the risk of potential in vivo brivanib alaninateeCCS binding, stance particle size. Tablets manufactured with drug substance
which could compromise bioavailability. Composition of the pro- particle size around the upper limit of the range typically produced
totype formulation is shown in Table 2. by the drug substance manufacturing process (D90 ¼ 133 mm) still

Figure 4. (a) Interaction plot of magnesium stearate and hydroxypropyl cellulose on tablet picking. (b) Contour plot of the effect of magnesium stearate and hydroxypropyl cellulose
on sticking to compression tooling.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 173

wet massing time. A 23 full-factorial design with 3 replicate center


points was used to study the effect of these parameters on tablet
dissolution at 2 kg scale in a 10-L high-shear granulator using a
single batch of drug substance. Details of study design and results
are reported elsewhere.16 Regression analysis showed that all main
effects of the 3 granulation parameters and 2-way interactions
were statistically significant (p < 0.05) for the amount of drug
dissolved at 30 min. Regression model fit to the DoE data and model
prediction for in-process material attributes and tablet dissolution
are shown in Figure 7. All 3 wet granulation process parameters
affected tablet dissolution in the same direction. Higher level of
water, impeller speed, or wet massing time resulted in a decrease in
dissolution rate. Tablet dissolution rate increased with the increase
in granule pore diameter (determined by MIP; Fig. 8). In addition,
Figure 5. Material attributes and process parameters impacting tablet dissolution. granule pore diameter showed similar dependence on granulation
parameters as tablet dissolution rate (Fig. 7b), suggesting that the
effect of granulation parameters on tablet dissolution is achieved
met dissolution limit (Fig. 6). However, dissolution of tablets
through their impact on granule porosity. In contrast, dissolution
manufactured with this large drug substance particle size was
showed poor correlation with granule size (mean granule diam-
lower compared with the smaller drug substance particle size (%
eter; Fig. 8). In addition, tablet disintegration did not correlate with
dissolved at 30 min was 89% and 99% for drug substance D90 ¼ 133
granulation parameters or tablet dissolution rate. This indicated
mm and 30 mm, respectively). Hence, drug substance particle size
that granule disintegration (rather than tablet disintegration) is the
was considered a CMA as variation in particle size was reflected in a
rate controlling step for dissolution. Hence, the 3 wet granulation
change in the tablet dissolution rate.
parameters highlighted in Figure 5 were considered critical for
Binder (HPC) particle size and solution viscosity, disintegrant
dissolution through their impact on granule porosity. These data
(CCS) particle size, and lubricant (magnesium stearate) surface area
are consistent with recent studies highlighting that granule
were also evaluated for their potential impact on tablet dissolution
porosity is the overriding mechanism impacting drug product
rate. This was achieved by using alternate vendors for these ex-
patient-centric CQAs such as drug release (dissolution)17 and
cipients exhibiting differences in the above attributes of interest.
bioavailability. Efforts have been made to study the kinetics of
Variation in these excipient attributes resulted in minimal change
granule consolidation during wet granulation and to make real-
in dissolution rate, and hence they were considered noncritical
time measurements of granule densification during wet granula-
(Fig. 5).
tion processes.18 These reports and our present study indicate that
particle size alone may not be an adequate response parameter to
Process Parameters and Dissolution. Based on risk assessment, assess granulation outcomes.19,20
Milling Unit Operation. A milling study was conducted to eval-
development studies were conducted to assess the impact of pro-
cess parameters in the following unit operations on tablet disso- uate 3 milling parameters: (1) screen size for wet milling, (2) screen
size for dry milling, and (3) impeller speed for dry milling. A split-
lution rate: wet granulation, wet milling, dry milling, and
lubrication. plot DoE design was used for the study by making a single wet
granulation batch, which was split into 2 sublots for wet milling;
Wet Granulation Unit Operation. A preliminary screening study
was conducted to select wet granulation parameters to be studied each was then used for a full-factorial study of the 2 dry milling
parameters. There was no impact of the change in milling param-
in a full-factorial DoE study. The results of the screening study
identified the following process parameters for the granulation eters on tablet dissolution despite the small changes observed in
the granule particle size distribution resulting from the different
process DoE study: (1) water amount, (2) impeller tip speed, and (3)
milling conditions. This is consistent with the findings from the wet
granulation study, which concluded that granule size does not
impact tablet dissolution rate. Milling parameters were, therefore,
designated as noncritical with respect to tablet dissolution as
shown in Figure 5.
Blending (Lubrication) Unit Operation. A magnesium stearate
blending (lubrication) study was carried out to assess the risk of
overblending of magnesium stearate on tablet dissolution. Increasing
blending time of magnesium stearate is known to reduce tablet
dissolution rate in some cases on account of the increased shearing of
magnesium stearate layers, which subsequently coat and impart
more hydrophobic characteristics to granule surfaces (a phenomenon
commonly referred to as overlubrication).21-23 In the manufacturing
process for brivanib alaninate tablets, magnesium stearate is blended
for 250 revolutions in a bin blender. Increasing number of revolutions
to 1000 did not have any impact on tablet dissolution. To further
assess the risk of overlubrication, the concentration of magnesium
stearate was also increased to 1.5% w/w (from 1.25% w/w in the
selected formulation) and also blended for 1000 revolutions. These
Figure 6. Dissolution profiles of brivanib alaninate tablets manufactured using drug
substance with different particle size. Dissolution in 50-mM phosphate buffer at pH 6.8
changes did not impact tablet dissolution which confirmed low risk of
with 1% Triton-X; USP Apparatus II; paddle speed of 75 rpm. Error bar represents overlubrication. Magnesium stearate blending time (number of
1 standard deviation. blender revolutions) was, hence, designated as noncritical.
174 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

Figure 7. Regression model for the effect of wet granulation process parameters on in-process material and tablet attributes. (a) Model fit to DoE data. (b) Model prediction;
figure represents main effects plots with 95% confidence intervals, showing the change in response as the variable is changed from the low to the high levels while holding other
variables at their center point value.

Tablet dissolution was also found to be independent of tablet


hardness (data not shown), which was not unexpected considering
that the rate limiting step of tablet dissolution is granule disinte-
gration rather than tablet disintegration as described previously.

Figure 8. Correlation of granulation particle size (GMD) and granule mean pore Figure 9. Material attributes and process parameters impacting water content and
diameter with tablet dissolution. impurities.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 175

that BMS-540215 formation on stability is dependent on initial


tablet water content, particularly for the aluminum blister package.
The initial water content in the tablets was, therefore, designated as
a CQA.
Control of water in the final product is achieved by (1) drying of
the granulation and tablets in the fluid bed and coating unit op-
erations, respectively; (2) bulk storage of the tablets in sealed
aluminum bags with desiccants; (3) final packaging of the tablets
under controlled humidity conditions; and (4) package design to
control water ingress into the package on shelf life. Water added
during wet granulation is removed by the fluid-bed drying unit
operation to an in-process limit aligned with the target water limit
of the tablets at release. An in-line NIR method was developed for
the determination of granulation water content in the fluid-bed
dryer. Subsequent to wet granulation and fluid-bed drying, mois-
ture pick-up by the product can take place during milling, blending,
Figure 10. Material attributes and process parameters impacting potency and content
uniformity. compression, and coating unit operations. Moisture pick-up could
be avoided by humidity control of the manufacturing environment.
Alternatively, tablet drying in the coating unit operation can be
Water Content and Impurities used to remove any water absorbed during process steps subse-
Brivanib alaninate is an ester prodrug which undergoes hydro- quent to fluid-bed drying. The latter strategy was implemented to
lysis reaction resulting in the formation of parent compound BMS- obviate the need for strict humidity control of the manufacturing
540215 (brivanib).24 BMS-540215 is the only degradant observed in environment. An initial drying step of the core tablets was used to
the drug product on accelerated and long-term stability studies. ensure acceptable core tablet moisture control before initiation of
Hence, this degradant is the major determinant of product shelf life. the coating process. This was considered necessary as drying of
The hydrolysis reaction of brivanib alaninate involves water as a tablet core after coating is applied becomes kinetically more diffi-
reactant. Thus, the rate of formation of BMS-540215 was found to cult. In addition, a drying step after the application of the film coat
be a function of relative humidity (water activity) and the storage was also implemented to remove any additional moisture pick-up
temperature.25 during coating and to ensure moisture control of the final coated
Process parameters and material attributes studied based on tablet. Both drying steps were controlled by a tablet LOD method.
risk assessment for water content and impurities are shown in Subsequent to film coating, tablet water content was controlled by
Figure 9. Relative humidity inside the package on stability is a the use of protective (moisture impermeable) bulk package and
function of initial tablet water content and package design (mois- control of humidity in the packaging environment.
ture permeation rate through the pack and the amount of desiccant
in the package, if present). A mathematical model was developed Degradation During Processing. Because BMS-540215 is formed by a
and previously reported25 which quantitatively predicts the effect hydrolytic reaction, the risk presented by aqueous wet granulation
of tablet initial water content and package parameters on the rate to degradation was extensively assessed. Preliminary studies using
of formation of BMS-540215 on stability. The concentration of BMS- target wet granulation parameters showed no increase in the level
540215 versus time profile was simulated using a kinetic model for of BMS-540215 (or any other impurity) during wet granulation
the formation of BMS-540215 as a function of relative humidity in even when the wet mass was stored for 25 h at room temperature
the environment. The kinetic model was combined with a sorption- before drying. In addition, the effect of wet granulation process
desorption moisture transfer model which calculates the relative parameters on the formation of BMS-540215 during wet granula-
humidity inside the package at different time points. The combined tion was also assessed. Granulation conditions imparting high
model, which simulates the amount of BMS-540215 in the pack- levels of water and shear forces were considered potential risk
aged tablets as a function of time, was used to study the effect of factors for brivanib alaninate hydrolysis. Therefore, batches man-
different variables such as package permeability and initial tablet ufactured using the high levels of water, impeller speed, and wet
water content on product stability. Model simulations indicated massing time (in the DoE study described previously) were tested
for their impurity content. No increase in any impurity level was
observed in these batches compared with the drug substance
indicating that wet granulation process parameters are not critical
for BMS-540215 formation in the tablet. The low risk presented by
wet granulation to tablet impurity content is attributed to the low
solubility of brivanib alaninate in the granulating liquid (water).
Initial impurity level in the tablet is hence determined solely by its
level in the drug substance used in manufacturing. Impurity level in
the drug substance, including BMS-540215, is therefore considered
CMA and is controlled by the drug substance specifications.

Potency and Content Uniformity


Process parameters and material attributes studied based on
risk assessment for potency and content uniformity are shown in
Figure 10.
Blending parameters and drug substance particle size presented
low risk on account of the high drug loading of brivanib alaninate in
Figure 11. Material attributes and process parameters impacting tablet appearance. the tablets (50% w/w). Therefore, they were not studied for their
176 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

impact on tablet potency and content uniformity. For high drug Tablet erosion during coating is a typical risk to the appearance
load formulations, potency and content uniformity are typically of a film-coated product and hence presents a risk to the appear-
controlled primarily by average tablet weight and weight variation. ance of brivanib alaninate tablets. Tablet hardness was therefore
These were, hence, considered critical for the control of these CQAs. controlled to minimize erosion risk. A lower limit of tablet hardness
In-process material attributes known to impact tablet weight was identified which showed low friability in the USP test (<0.2%)
variation during compression includes granulation particle size and resulted in no erosion during tablet coating. Analysis of
distribution and flow properties. Wet granulation and milling compaction data from the wet granulation DOE study revealed no
process parameters were evaluated for their effect on granulation significant main effects of granulation parameters with respect to
particle size distribution, flow properties, and tablet weight varia- compaction responses. All batches in the study had comparable
tion in compression in the abovementioned DoE studies. Although compaction profiles indicating that variation in granulation pa-
change in wet granulation and milling parameters caused some rameters had no impact on the compaction properties of the final
variation in final blend particle size distribution and flow rate blend. Moreover, the maximum hardness achieved in the study for
(measured by an Erweka® flow tester; Fig. 7), this variation in all batches was well above the target tablet hardness.
granule size distribution and flow rate had no impact on tablet
weight variation in compression studies. Wet granulation and
Control Strategy
milling parameters are hence considered noncritical with respect
to their impact on potency and content uniformity.
Based on development studies and knowledge of CPPs and at-
tributes impacting CQAs, a combination of control elements were
Tablet Appearance implemented to ensure that the drug product met the required
Picking and sticking tendency during tablet compression caused quality standards. The drug product manufacturing control strategy
by inherent properties of the brivanib alaninate drug substance was included (1) process parameter controls (process design space), (2)
identified early in development as a risk to tablet appearance and to input material controls, (3) in-process attribute controls during
process yield and cycle time.14 The wet granulation process was manufacturing, and (4) end-product testing. The control strategy is
primarily selected to manufacture brivanib alaninate tablets to essentially a combination of limits on all critical starting material
mitigate sticking risk. Incorporation of the drug substance particles attributes, in-process material attributes, and process parameters
in granules manufactured by wet granulation was shown to elim- identified for each CQA (Figs. 5, 9, 10, and 11). If a parameter or
inate sticking.14 Moreover, the concentration of the binder (HPC) attribute is critical for >1 CQA, the limit in the control strategy is
was found to be statistically significant with respect to the sticking based on the CQA with the tightest requirement.
response in the formulation DoE study and hence was optimized in In addition, the control strategy also included at-line release
the selected formulation to eliminate sticking as described previ- tests and finished product release tests. The at-line release tests
ously. HPC reduces sticking by reducing fines and ungranulated included testing of core tablets for potency and uniformity of
drug substance particles in the final blend. In addition to HPC, the dosage units. Release tests of the finished product (film-coated
concentration of the lubricant (magnesium stearate) was found to tablets) include dissolution, water content, appearance, and iden-
be statistically significant in the formulation DoE and was similarly tification. The control strategy is listed in Tables 3 and 4 and
optimized to mitigate the sticking risk. described in the following sections.
Based on the understanding of the sticking risk, drug substance
particle size, HPC and magnesium stearate material attributes, and
Process Parameters Design Space
wet granulation, milling and lubrication process parameters were
Wet granulation parameters (water amount, impeller speed and
studied for their effect on sticking (Fig. 11).
wet massing time) were the only process parameters identified as
critical based on the studies described previously. Wet milling, dry
Material Attributes and Sticking. The effect of drug substance par- milling, and lubrication parameters studied were found to be
ticle size on sticking was evaluated. Small drug substance particle noncritical as all CQAs showed little sensitivity to these parameters.
size at the lower specification limit did not show sticking when The results of the wet granulation DoE study described previ-
used for drug product manufacturing. Variation in HPC particle size ously formed the basis for assigning the 3 wet granulation pa-
and viscosity and magnesium stearate surface area were also rameters as critical parameters on account of their impact on tablet
studied for potential effect on tablet sticking during compression. dissolution rate. Because all batches from the DoE study met the
As mentioned previously, this was accomplished by using alternate acceptance criteria for dissolution (not less than 75% dissolved in 30
vendors for these excipients exhibiting differences in the attributes min) and all other CQAs, parameter ranges in the DoE are consid-
of interest. No effect was observed due to variation in these ered multivariate PARs and hence represent the process design
excipient properties, and all batches made with the different space (Table 3). Parameter values for the center point batches were
excipient properties showed no sticking during compression. established as the target parameters.
The design space for the wet granulation parameters was
Process Parameters and Sticking. None of the wet granulation or established based on the DoE study at 2.5-kg scale. According to the
milling DOE batches described previously showed any sign of International Conference on Harmonization guideline, a design
sticking to the compression tooling. Similarly, the shorter blending space can be developed at any scale and be applicable to the
time of magnesium stearate did not result in any sticking in the commercial scale if the design space is shown to be scale
lubrication study.
Based on these results, no parameters or attributes were found
Table 3
to be critical for sticking. The selection of the manufacturing pro- Control Strategy: Process Parameters Design Space
cess (wet granulation) and the optimization of HPC and magnesium
Process Parameter Unit Target (Design Space Range)
stearate levels in the selected formulation resulted in a robust
formulation that showed no sensitivity to process parameters or Water for granulation % w/w 46.5 (44-49)
input material attributes with respect to blend sticking tendency Impeller tip speed m/s 4.8 (3.6-6.0)
Wet massing time s 30 (10-50)
during compression.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 177

Figure 12. Comparison of brivanib alaninate in-process material and tablet attributes for product manufactured at 3 different scales. (a) Particle size distribution of granulation,
(b) compaction profile of final blend, and (c) tablet dissolution profile.

independent.26 As a result, a strategy to justify the extrapolation of substance particle size was therefore necessary to ensure consistent
the design space from the small scale to the commercial scale was and acceptable dissolution of the tablets. A particle size specification
established. The strategy described below provides the necessary of 40-120 mm (D90) was established to ensure acceptable dissolution
justification, while minimizing the number of batches to be man- rate (not less than 75% dissolved at 30 min). While the upper limit is
ufactured at the commercial scale. established to ensure acceptable dissolution, the lower limit of par-
A prerequisite for the extrapolation among scales is to have the ticle size was set to minimize risk of sticking to compression tooling
design space expressed in scale-independent terms. The critical resulting from fine drug substance particles.
wet granulation parameters of the brivanib alaninate design space Because both wet granulation parameters and drug substance
are expressed in scale-independent terms as shown in Table 3. The particle size are critical to dissolution rate, it was necessary to
suitability of the scale-independent terms used for the design space establish that the upper limit of the drug substance particle size
was verified by comparing product quality attributes at 3 scales, range shows acceptable tablet dissolution even at the edge of the
representing laboratory scale, pilot scale, and commercial scale. Key process design space. Because the wet granulation DoE was con-
attributes of these batches manufactured at the 3 scales using ducted with drug substance having D90 of 68 mm, additional study
target parameters values were comparable as shown in Figure 12. was necessary to demonstrate performance of the larger particle
In addition, the design space was also verified by manufacturing size at the edge of the process design space. A risk-based approach
2 batches at 2 extreme regions in the design space at the com- was used to achieve this purpose leveraging the knowledge
mercial scale (off-target batches). The strategy for selecting the established from the wet granulation study which illustrated that
parameters for those batches was based on selection of worst-case combination of the high levels of all 3 parameter values results in
combination of the 3 critical parameters. Because the 3 critical the lowest dissolution rate. Hence, the worst-case combination of
granulation parameters affect dissolution in the same direction granulation parameters which was identified from the DoE study
(Fig. 7b), combination of the high levels of all 3 parameters repre- (high water level, high impeller speed and high wet massing time)
sents the highest risk for slow dissolution rate. Conversely, com- was evaluated in conjunction with large drug substance particle
bination of the low levels of all parameters represents the highest size (D90 ¼ 106 mm) to ensure that the worst-case combination of
risk of “undergranulation” and associated risks of poor flow prop- process parameters and drug substance particle size would result in
erties and sticking to tablet compression tooling. Results from the 2 an acceptable dissolution rate. As shown in Figure 13, dissolution of
off-target batches met all acceptance criteria, hence providing tablets manufactured with this worst-case combination of process
additional justification for the applicability of the design space to parameters and particle size showed acceptable dissolution profile
the commercial scale. which met the acceptance criteria.
The other critical input material attribute (drug substance im-
Input (Raw) Material Controls purity content) was controlled by incoming material specifications,
Drug substance particle size and impurity content were the only with a limit of not more than 1.0% on the parent compound (BMS-
input material attributes identified as critical to product perfor- 540215).
mance or CQAs. None of the excipient-related attributes was found
to be critical as concluded from the development studies. A corre- In-Process Attributes Control
lation between the particle size of the drug substance and dissolu- Fluid-Bed Drying. Water content of the granules during drying was
tion rate was established as previously discussed. Control of the drug tested and controlled by the NIR spectroscopy method to a limit of
178 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

Figure 13. Dissolution profile of tablets manufactured with large drug substance
particle size and high level of wet granulation parameters (water amount, impeller
speed, and wet massing time). Dissolution in 50-mM phosphate buffer at pH 6.8 with
1% Triton-X; USP Apparatus II; paddle speed of 75 rpm.

not more than 1.3% w/w. The limit was implemented to ensure that
water added during wet granulation is removed by drying in the
fluid-bed dryer and to account for moisture associated with the
extragranular excipients added downstream. In addition, the limit
was set to enable the coated tablet to meet water content accep-
tance limit in the final product.
The established NIR method was used to monitor granulation
moisture during fluid-bed drying of several development and
commercial-scale batches. Comparison between the NIR and LOD
results showed close agreement (Fig. 14), and it was determined
that moisture levels could be accurately measured between 0% and
3% with approximately 0.25% resolution. The NIR method suitably
monitored the drying process of commercial-scale batches at the
manufacturing site, enabling determination of the drying end
Figure 15. (a) NIR calibration curve for at-line measurement of core tablet potency.
point. Predicted tablet assay by NIR method, reference assay by HPLC. (b) Potency results for
full-scale brivanib tablet batches comparing composite HPLC (for coated tablets) versus
at-line NIR (for core tablets). “Minimum Tablet” and “Maximum Tablet” are for the NIR
Compression. As typically implemented in tablet compression, up- potency determination.
per and lower acceptance limits for core tablet weight (both indi-
vidual and mean tablet weight) were established to ensure
implemented to ensure that tablets could withstand the coating
acceptable assay and content uniformity of the core tablets. Simi-
process, packaging, and shipping.
larly, control of tablet hardness within the acceptance limits was

Film Coating. As mentioned previously, tablet-coating process was


designed to control the water level in the core tablets and to
remove any additional water picked up by the drug product during
film coating of the tablets. To achieve these objectives, the coating
process was designed to include 3 stages: (1) drying of core tablets,
(2) application of film coat (spray), and (3) drying of the coated
tablets. Water content of uncoated tablets (before the start of
coating) and coated tablets (after the end of coating) was deter-
mined by LOD method and controlled by an in-process specification
limit. Water content limit in the core tablet was established at not
more than 1.6% w/w. The control on the water level in the core
tablets ensured that any water picked up by the product during the
stages of manufacture after fluid-bed drying was removed before
coating.
A limit on the water level for the coated tablet was also set at not
more than 1.8% w/w to ensure that any water picked up by the
tablets during coating was removed. Effectiveness of this coating
approach to achieve acceptably low water level in the coated tab-
lets was established and previously reported.25 Water limit of the
Figure 14. Comparison of online NIR and at-line LOD for moisture monitoring during coated tablets was based on the simulation of the effect of initial
fluid-bed drying of brivanib alaninate granulation. water content on the rate of hydrolysis of brivanib alaninate. Using
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 179

Table 4
Control Strategy: Raw Materials, In Process Controls and Release Tests

Tablet Quality Attribute Raw Material In-Process Control At-line Release Test Off-line Release Test

Drug release Drug substance particle size Dissolution


Impurities Drug substance impurities
Water content Water content at end of fluid-bed
drying of granules (NIR, NMT 1.3%)
Water content of preheated core
tablets in coating pan (LOD, NMT 1.6%)
Water content of tablets at end of
coating (LOD, NMT 1.8%)
Potency Tablet weight during compression Core tablet potency by NIR
Content uniformity Tablet weight during compression Uniformity of dosage units by
weight variation on core tablets
Appearance Tablet hardness Physical imperfections AQL

AQL, acceptable quality limit of tablet defects; NMT, not more than.

the predictive model mentioned previously, the formation of obviates the need for testing of impurities for tablet release. Im-
BMS-540215 as a function of the initial tablet water content was purities are tested on market life stability as BMS-540215 level
examined, and results identified an acceptable water content increases over-shelf life.
release limit of not more than 2.4% w/w for final coated tablets. The
1.8% in-process limit of the coated tablets is tighter than release Discussion
limit to account for the variability of the in-process LOD method
and the KarleFischer release test method and ensures that tablets An integrated and overarching application of QbD principles to
meet the specification limit for water content on release testing.25 modern drug product development requires a holistic under-
standing of the effect of input variables which include (1) raw
Product Release Testing material properties and their potential variability, (2) formulation
At-Line Release Tests composition (qualitative and quantitative), and (3) manufacturing
Tablet Potency. An at-line release testing for tablet potency is process parameters. In this article, a drug product control strategy
performed on the core tablets by the NIR method. Tablet samples was developed which encompasses both material property and
were collected throughout compression and used for the potency process parameter controls, with appropriate in-process and
analysis. Tablet potency by the NIR method must meet the potency finished product analytical tests and specification limits (Tables 3
limit on the release specification. No potency release testing is and 4). These analytical tests encompass both conventional
performed on the final coated tablets as the potency does not analytical tests and process analytical technology. In the following
change during film coating. Data from development and technol- section, some of the key elements from the brivanib alaninate QbD
ogy transfer batches confirmed this requirement. development are discussed further.
Figure 15a shows the brivanib alaninate tablet calibration curve
for the NIR method with 7 concentration levels of brivanib alani- Building Process Robustness by Formulation Design
nate and 17 combinations of excipient concentration levels. The
calibration root mean squared error of approximately 1.6% w/w and It should be recognized that building manufacturing process
a correlation coefficient of 0.99 demonstrated strong correlations robustness starts with appropriate formulation design. Traditional
between the HPLC measurements and NIR predictions. A compar- product development efforts focus on sequential development of
ison between the HPLC composite potency results and the NIR formulation composition, manufacturing process, and drug product
potency results was performed for production of full-scale batches control strategy. Because excipients affect process performance,
of brivanib alaninate tablets. The results, plotted in Figure 15b, selection of formulation composition without consideration of the
show excellent agreement between NIR and HPLC results, process can limit process robustness. Process development efforts
demonstrating the capability of the NIR method to quantitate bri- aimed at establishing parameter space where process performance
vanib alaninate level in the core tablets. is minimally impacted by sources of variability would be signifi-
Tablet Content Uniformity. Tablet uniformity was also tested on cantly hampered by suboptimal formulation composition. In the
the core tablets using a weight variation method as allowed by the integrated development exemplified in this article, the assessment
compendia at the high drug loading (50% w/w). of robustness was studied across the unit operations at the
Final Product Off-Line Release Tests. Off-line release tests are formulation design stage through fewer, mechanistically driven
implemented for dissolution and tablet identity (Raman spectros- experiments.
copy method) and must conform to the acceptance criteria of the As an example of how process robustness can be built into
release specifications. In addition, a water content release test by a excipient selection and formulation design, we studied the effect of
KarleFischer method is implemented with a limit of not more than the disintegrant, CCS versus CPVP, on the end point of the wet
2.4% w/w as described previously. granulation process. Performance of the wet granulation process
No final product release test was proposed for impurities. This is with both disintegrants was evaluated and CCS was selected as the
based on product understanding and input material controls in intragranular disintegrant on account of the greater robustness of
place. BMS-540215 is the only degradant of the drug product. the CCS formulation with respect to granulation end point.
However, BMS-540215 increases only on shelf life and does not Another example was provided by the formulation optimization
change during drug product manufacturing, even at the worst-case to enhance its performance in the tablet compression process with
extremes of the granulation parameter design space. Therefore, respect to sticking to compression tooling. As a key risk to process
controlling the level of BMS-540215 in the drug substance ensures robustness, the effect of formulation composition on sticking po-
acceptable degradant level in the drug product at release and tential was studied in the formulation-ruggedness DoE. Findings
180 S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181

related to the role of binder and lubricant and their interaction on demonstrated by manufacturing product at 3 different scales (form
sticking were the basis for the optimization of the concentration of pilot to production scales) using target parameters; key product
HPC and magnesium stearate in the final formulation. The opti- attributes were shown to be similar across the 3 scales; (3) pro-
mized formulation demonstrated excellent robustness and showed duction scale batches were manufactured at 2 points in the process
no sticking in the process development studies across the full range parameter space which represent worse case parameter combina-
of parameters tested. tions with respect to CQAs. Both batches showed acceptable attri-
butes. The approach used was hence based on process
Design Space Strategy understanding established from the development studies and was
successful in minimizing number of batches manufactured at the
An integrated design space that incorporates a range of production scale.
permissible or expected variability in material properties and
process variables presents significant logistic challenge. These Model-Based Specification Limits
challenges stem from the wide range of input variables and re-
sponses that must be evaluated. The following 2 strategies can be Final product and in-process specification limits are key com-
adopted to address these challenges: (1) generate an extended ponents of the control strategy. Specifications need to be linked to 1
experimental space using multifactorial statistical DoE study or (2) or more CQA with limits established within which product per-
adopt a selective and sequential study designs of which parameters formance is acceptable and should be established based on un-
and material attributes are evaluated based on mechanistic un- derstanding of the factors affecting the CQA. Traditionally,
derstanding of their effect on product attributes. specification limits are based on data of key batches (e.g., long-term
A single DoE that incorporates variables across material prop- stability and clinical batches used in pivotal trials) and perceived
erties and process parameters collectively, although feasible, be- process capability. Such an approach is not very effective because
comes prohibitive in material, time, and effort requirements.27 To there are usually other factors affecting a given CQA, in addition to
overcome the limitations of a single comprehensive study, we the attribute for which the specification limit is being set. Estab-
highlight in this article the alternative strategy of using a sequential lished limits using the traditional approach do not account for the
and selective DoE. Using this approach, studies do not necessarily multivariate effect of these factors on the CQA. Mathematical
use exhaustive statistical designs but are based on sound scientific modeling is a useful tool to quantify the effect of these factors on
and mechanistic understanding of underlying causes of product CQAs and provides the basis for product and in-process specifica-
responses. For example, the effects of wet granulation process pa- tions. For brivanib alaninate tablets, degradant formation depends
rameters and material properties on the dissolution rate of brivanib on initial water content of tablets and on packaging factors such as
alaninate tablets were evaluated in separate studies. CPPs, their package permeability and presence of desiccant. A model-based
design space, and the mechanism of their effect on tablet dissolu- approach was used to predict the effect of these different factors
tion were established in the process DoE. Material properties study on rate of degradant formation and to rationally establish a speci-
identified drug substance particle size as the critical property for fication limit on initial tablet water content. The model quantita-
dissolution and established an acceptable particle size range. tively predicted the effect of the different variables on degradation
Because the wet granulation study used target drug substance rate. Hence, the model allowed for the initial tablet water content
particle size, while the particle size study was conducted at target limit to be set based on an in silico study of the variable space
wet granulation parameters, combination of the 2 acceptable instead of relying on limited set of batch data.
ranges from the 2 studies required further justification. Product and
process understanding established in the respective studies were Conclusions
leveraged to generate experimental space that captures the worst-
case combination of wet granulation parameters and drug sub- Identification of CQAs of brivanib alaninate tablets, risk assess-
stance particle size. Additional experimentation of this worst-case ment, and focused development studies were instrumental to the
space was conducted to confirm the multivariate acceptability of identification of an effective control strategy based on QbD prin-
the combination of acceptable ranges from the process and mate- ciples. Controls were implemented on input and in-process mate-
rial property studies. The application of such strategy, as demon- rials and on manufacturing process parameters, which ensure that
strated in this article, rationally reduces the extent of final product consistently meets criteria established for CQAs. This
experimentation required by leveraging the scientific and mecha- demonstration of QbD principles to integrated and holistic drug
nistic understanding of variables that impact product quality and product development highlights the value of mechanistically
enables holistic approach to the development of a control strategy driven experiments in building product and process robustness
for assuring product quality. while addressing key risks to the patient, manufacturing plant, and
The manufacturing scale used to establish design space presents product development.
another challenge. Ideally, design space would be established at the
commercial production scale. However, this strategy presents sig- References
nificant challenges associated with cost and availability of drug
substance. Limited availability of drug substance during drug 1. Yu LX. Pharmaceutical quality by design: product and process development,
product development stage makes this approach impractical in understanding, and control. Pharm Res. 2008;25(4):781-791.
2. Yu LX, Amidon G, Khan MA, et al. Understanding pharmaceutical quality by
many cases. ICH Q8 permits development of design space at pilot design. AAPS J. 2014;16(4):771-783.
scale and extrapolation to the production scale if properly justified. 3. Awotwe-Otoo D, Agarabi C, Wu GK, et al. Quality by design: impact of
However, the ICH guideline does not prescribe an approach for formulation variables and their interactions on quality attributes of a lyophi-
lized monoclonal antibody. Int J Pharm. 2012;438(1-2):167-175.
justifying extrapolation of design space from pilot scale. In this
4. Kona R, Fahmy RM, Claycamp G, Polli JE, Martinez M, Hoag SW. Quality-by-
article, an approach for the extrapolation of wet granulation design design III: application of near-infrared spectroscopy to monitor roller
space is used which consists of 3 components: (1) wet granulation compaction in-process and product quality attributes of immediate release
parameters at the pilot scale are expressed in scale-independent tablets. AAPS PharmSciTech. 2014;16(1):202-216.
5. Merritt JM, Viswanath SK, Stephenson GA. Implementing quality by design in
terms (impeller tip speed, ratio of water to solids and wet mass- pharmaceutical salt selection: a modeling approach to understanding dispro-
ing time); (2) scale independence of these parameters was portionation. Pharm Res. 2013;30(1):203-217.
S.I.F. Badawy et al. / Journal of Pharmaceutical Sciences 105 (2016) 168e181 181

6. Saurí J, Mill ~e
an D, Sun -Negre JM, et al. Quality by design approach to under- 17. Wolfe S, Pafiakis S, Tang D, et al. Effect of Tablet Pore Size Distribution on
stand the physicochemical phenomena involved in controlled release of Dissolution upon Accelerated Storage Conditions. 2013. AAPS Annual Meeting,
captopril SR matrix tablets. Int J Pharm. 2014;477(1-2):431-441. San Antonio, TX.
7. Visser JC, Dohmen WMC, Hinrichs WLJ, Breitkreutz J, Frijlink HW, 18. Narang AS, Sheverev VA, Stepaniuk V, et al. Real-time assessment of granule
Woerdenbag HJ. Quality by design approach for optimizing the formulation densification in high shear wet granulation and application to scale-up of a pla-
and physical properties of extemporaneously prepared orodispersible films. Int cebo and a brivanib alaninate formulation. J Pharm Sci. 2015;104(3):1019-1034.
J Pharm. 2015;485(1-2):70-76. 19. Pandey P, Sinko PD, Bindra DS, Hamey R, Gour S, Vema-Varapu C. Processing
8. Bohlin M, Jones H, Black S. New developments in scale-up and QbD to ensure challenges with solid dosage formulations containing vitamin E TPGS. Pharm
control over product quality. Am Pharm Rev. 2009;12(7):52-57. Dev Technol. 2013;18(1):296-304.
9. Kayrak-Talay D, Dale S, Wassgren C, Litster J. Quality by design for wet gran- 20. Pandey P, Tao J, Chaudhury A, Ramachandran R, Gao JZ, Bindra DS. A combined
ulation in pharmaceutical processing: assessing models for a priori design and experimental and modeling approach to study the effects of high-shear wet
scaling. Powder Technol. 2013;240:7-18. granulation process parameters on granule characteristics. Pharm Dev Technol.
10. Kushner J, Langdon BA, Hicks I, et al. A quality-by-design study for an 2013;18(1):210-224.
immediate-release tablet platform: examining the relative impact of active 21. Lerk CF, Bolhuis GK, Smallenbroek AJ, Zuurman K. Interaction of tablet dis-
pharmaceutical ingredient properties, processing methods, and excipient vari- integrants and magnesium stearate during mixing. II. Effect on dissolution rate.
ability on drug product quality attributes. J Pharm Sci. 2014;103(2):527-538. Pharm Acta Helv. 1982;57(10-11):282-286.
11. Prpich A, Am Ende MT, Katzschner T, Lubczyk V, Weyhers H, Bernhard G. AIChE 22. Pingali KC, Mendez R. Nanosmearing due to process sheardinfluence on
Annual Meeting, Conference Proceedings. 2008. powder and tablet properties. Adv Powder Technol. 2014;25(3):952-959.
12. Huynh H, Ngo VC, Fargnoli J, et al. Brivanib alaninate, a dual inhibitor of 23. Sheskey PJ, Robb RT, Moore RD, Boyce BM. Effects of lubricant level, method of
vascular endothelial growth factor receptor and fibroblast growth factor re- mixing, and duration of mixing on a controlled-release matrix tablet con-
ceptor tyrosine kinases, induces growth inhibition in mouse models of human taining hydroxypropyl methyl cellulose. Drug Dev Ind Pharm. 1995;21(19):
hepatocellular carcinoma. Clin Cancer Res. 2008;14(19):6146-6153. 2151-2165.
13. Narang AS, Badawy S, Ye Q, et al. Role of self-association and supersaturation in 24. Zhao F, Derbin G, Miller S, Badawy S, Hussain M. Acid-catalyzed hydrolysis of
oral absorption of a poorly soluble weakly basic drug. Pharm Res. 2015. Article BMS-582664: degradation product identification and mechanism elucidation.
in press; doi:10.1007/s11095-015-1645-y. J Pharm Sci. 2012;101(9):3526-3530.
14. Narang AS, Badawy SIF, Subramanian GA, LaMarche KR, Bindra DS, Rao VM. 25. Badawy SI, Lin J, Gokhale M, et al. Quality by design development of brivanib
High Drug Load Tablet Formulation of Brivanib Alaninate. 2012. WO/2012/ alaninate tablets: degradant and moisture control strategy. Int J Pharm.
097222. 2014;469(1):111-120.
15. Narang AS, Yamniuk AP, Zhang L, et al. Reversible and pH-dependent weak 26. International Conference on Harmonization of Technical Requirements for
drug-excipient binding does not affect oral bioavailability of high dose drugs. Registration of Pharmaceuticals for Human Use. ICH Harmonized Tripartite
J Pharm Pharmacol. 2012;64(4):553-565. Guideline, Pharmaceutical Development Q8(R2), Current Step 4 Version.
16. Badawy SI, Narang AS, Lamarche K, Subramanian G, Varia SA. Mechanistic basis 27. Narang A. Addressing excipient variability in formulation design and drug
for the effects of process parameters on quality attributes in high shear wet development. In: Narang A, Boddu S, eds. Excipient Applications in Formulation
granulation. Int J Pharm. 2012;439(1-2):324-333. Design and Drug Delivery. New York, NY: Springer; 2015.

You might also like