Вы находитесь на странице: 1из 20

Tablet Compression

Optimizing Tablet
Compression
Frederick J. Murray

A
A process optimization ll aspects of pharmaceutical manufacturing face in-
template provides a practical creased pressure to improve production efficiency and
approach for maximizing
uptime, while maintaining the highest standards for
output and product quality
of an existing tablet product quality. For tablet compression equipment,
compression process. many companies seek to increase productivity of the existing
equipment platform prior to considering additional equipment. A
structured optimization template provides a practical approach to
maximizing both output and product quality of an existing tablet
compression process.

Tablet weight control


The key process parameter for any tablet compression operation is
tablet weight control. Assuming uniform distribution of the blend,
the ability to hold precise tablet weight is an absolute requirement
to delivering the prescribed dosage of active material. Tablet weight
control is influenced by a number of factors including press speed,
flow properties of the granulation, filling depth, feeder paddle con-
figuration, and working lengths of the upper and lower punches.
Press speed. The press speed and pitch circle diameter of the tablet
press die table will determine the tangential velocity of the press
tools. The lower press tools pass under the feed frame to fill the dies,
and based on the length of the feeder opening and tangential velocity
of the tools, the feeder dwell time may be determined. It is obvious
that a longer feeder dwell-time will permit more time to fill the dies
SHUTTERWORX/GETTY IMAGES

and that there may be a critical speed limit where it is impossible to


achieve uniform die filling because the feeder dwell time is too short.
Frederick J. Murray is
Flow properties of the granulation. A product with robust flow proper-
President of KORSCH America
Inc., tel: 508.238.9080, fred. ties will fill the dies uniformly at high speeds. Products with mar-
murray@korschamerica.com.
ginal or poor flow properties need long feeder dwell times and often
26 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
Tablet Compression
require a modified feeder paddle configuration to in the tablet weight. Precision tablet weight control
fill the dies uniformly. thus mandates excellent press-tool tolerances and
Filling depth. All tablet presses use a volumetric a corresponding press-tool maintenance program.
fill to obtain the desired tablet weight. The fill Understanding the issues that impact tablet weight
depth (i.e., position of the lower punch in the die control and can cause tablet weight variability is cru-
to achieve the desired tablet weight) is determined cial to a successful process optimization effort.
by the shape of the tablet and the bulk density of
the material. A small tablet diameter with a deep Tablet hardness
filling depth is obviously more difficult to fill than In addition to tablet weight, thickness and hard-
a larger tablet diameter with a small filling depth. ness are key quality parameters for tablet produc-
The ability to fill dies with deep filling depths can tion, and these attributes can be measured in real
certainly be rate limiting. time as the tablet press is producing tablets. The
Feeder paddle configuration. Most modern tablet tablet hardness will determine the dissolution rate,
presses use variable-speed power feeders with ro- which is crucial to ensure effective drug delivery.
tating feeder paddles that assist the transfer of ma- The tablet hardness is a function of the volume of
terial into the die. Feeder paddles generally come material in the die, and the magnitude and rate of
in a rectangular profile; however, many tablet compression force applied to the tablet. In most
presses are available with alternate feeder paddle cases, increasing the press force applied to the
designs, including round profiles and beveled pro- tablet will increase the corresponding tablet hard-
files. There is no real handbook to define the best ness. Some products, however, have a maximum
feeder paddle for any given product, and empirical hardness threshold, and higher press forces actu-
testing, such as described in this article, is required ally cause the tablet hardness to be reduced. Un-
to identify the optimal paddle configuration. derstanding the relationship between press force
Upper- and lower-punch working lengths. The role of and tablet hardness is a crucial component to any
press-tool working lengths on tablet weight control process optimization study.
is often overlooked. Variability in the lower-punch The rate that press force is applied to the tablet
working length will directly impact the volume of is a function of the tangential velocity of the press
material in each die and can, in itself, cause tablet tools, the diameter of the compression rollers, and
weight variation. Variability in the upper-punch the geometry of the press tool, and it is generally
working length will not directly impact the volu- referred to as compression dwell time. In simple
metric fill; however, because most modern tablet terms, the compression process imparts energy
presses are using press-force control systems as the into the tablet by applying a force over a period of
basis for tablet weight control and because upper time. If the time is reduced (when the press speed
punch working length variability will alter the tab- is increased), then the force must be increased
let thickness (and therefore, the press force), the to impart the same amount of energy. The press
force control system is now reacting to tolerance is- speed is thus another dimension to the press force
sues in the upper punches and not actual variability tablet hardness relationship.
28 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
Process optimization Figure 1: Tablet weight variation shown as relative standard deviation (Srel%) of individual tablet
template weight as a function of press speed.

With this fundamental 6%

understanding of the 5%
key process parameters
4%
and those factors that
Weight Srel %
3%
can impact quality pa-
2%
rameters, one can use
1%
the process optimization
0%
template, which consists
20 30 40 50 60 70 80 90
of the following four sets
Press speed (RPM)
of empirical testing.
Press speed/tablet qual-
ity. This testing consists Figure 2: Tablet thickness variation shown as relative standard deviation (Srel%) of individual
tablet thickness as a function of press speed.
of making the product to
6%
specification (i.e., target
5%
weight, thickness, and
hardness) and then mea- 4%
Thickness Srel %

suring the standard devi- 3%

ation of individual tablet 2%

weight, thickness, and 1%

hardness values across 0%


20 30 40 50 60 70 80 90
a defined speed range.
Graphs of standard de- Press speed (RPM)

viation of individual tab-


let weight, thickness, and Figure 3: Tablet hardness variation shown as relative standard deviation (Srel%) of individual
hardness variation versus tablet hardness as a function of press speed.

press speed will clearly 40%

define the press speed 35%

30%
range in which quality
25%
Hardness Srel %

tablets can be produced. 20%


Figure 1 plots the rela- 15%

tive standard deviation 10%

5%
(Srel %) of individual tab-
0%
let weight as a function of 20 30 40 50 60 70 80 90

press speed. In this ex- Press speed (RPM)


ample, there is a clear in-
Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 29
Tablet Compression
crease in the tablet weight Figure 4: Tablet hardness vs. compression force at different press speeds (i.e., compression
variability at press speeds dwell times).

higher than 60 RPM, thus 12

60 RPM is the maximum 10

Hardness (KP)
press speed given the flow 8

properties of the material 6

4
tested. Better flow prop-
2
erties would likely permit
0
higher press speeds. 2 4 6 8 10
30 RPM 45 RPM 60 RPM 75 RPM 90 RPM
Figure 2 plots the rela- Main compression force (kN)
tive standard deviation
(Srel %) of individual tab-
let thickness as a function of press speed and shows (i.e., compression dwell time) and will confirm
consistent control across the press speed range. the press force value that corresponds to the de-
Figure 3 plots the standard deviation (Srel %) of sired tablet hardness.
individual tablet hardness as a function of press The representative graph shown in Figure 4 indi-
speed. These data mirror the tablet weight data, cates that the compression dwell time does impact
with consistent control up to the 60 RPM press the tablet hardness, especially at the higher press
speed level. force levels. At 90 RPM, the tablet hardness is, on
The process capability (Cp) may be calculated average, 80% lower than the same tablet produced
for tablet weight, thickness, and hardness at each at the 30 RPM press speed level. Based on required
press speed using Equation 1. tablet hardness range, the expected press force
range can be easily determined.
Cp = (USL LSL) / 6 * [Eq. 1] Feeder speed/feeder paddle optimization. This test
consists of running the press at different press
Where Cp is process capability index, USL is speeds and adjusting the feeder speed across the
upper specification limit, LSL is lower specifica- range, while recording the relative standard de-
tion limit, and is standard deviation. viation of individual tablet weights. This measure-
Nominal process capability values of 1.33 or 1.50, ment is performed at each press speed and with
or higher, are generally indicative of a process that different feeder paddles designs (standard, round,
is under control. beveled) to determine the optimal feeder speed
Tablet hardness/compression force. This testing and feeder-paddle configuration.
consists of running the press at different com- If the results show that the standard deviation of
pression forces and measuring tablet hardness at tablet weight is not impacted significantly by the
the different press-speed levels. The force versus feeder speed, then it makes sense to run the feeder
hardness plot will confirm the ability to achieve speed at the lowest value to avoid overmixing or
the desired tablet hardness at each press speed shearing the granulation in the feeder.
30 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
M
ic S
ro pe
en ci
ca ali
ps sts
ul
at
io
n
Does your current CDMO
leave you with blurred vision?

6RPH&RQWUDFW'HYHORSPHQW0DQXIDFWXULQJ2UJDQL]DWLRQV &'02 JHW\RX


in the door then seem to go in multiple directions. There is no clear direction;
and the invoices are piling up.
At Coating Place our experienced staff believes the best results come with a
clear understanding of your ideas and goals. Trust the knowledge, quality and
LQVLJKW&RDWLQJ3ODFHSURYLGHVWRJHW\RXUYLVLRQLQWRIRFXVDQGGHOLYHUHG

A UH
DQGVRPXFKPR

EMAILINFO COATINGPLACECOMs  s#OATING0LACE )NC "OX 6ERONA 7ISCONSIN53!sCOATINGPLACECOM


LQIR#FRDWLQJSODFHFRP  
Tablet Compression
lock in the optimal feeder
Figure 5: Weight variation shown as relative standard deviation (Srel%) vs. feeder speed at a series
of press speeds (3090 RPM indicated as different colors) using a rectangular feeder paddle. speed at each press speed.
Figure 7 shows the im-
6%
pact of the feeder-paddle
5%
configuration on the rela-
4%
tive standard deviation of
Weight Srel %

3%

2%
individual tablet weight at
1% a variety of different press
0% speeds and feeder speeds.
10 20 30 40 50 60
The results show compa-
30 RPM 45 RPM 60 RPM 75 RPM 90 RPM rable performance of the
Feeder speed (RPM), rectangular paddles two paddles at the 30 RPM
press speed (see Figure 7a).
Figure 6: Weight variation shown as relative standard deviation (Srel%) vs. feeder speed at a
At the 60 RPM press speed,
series of press speeds (3090 RPM indicated as different colors) using a round feeder paddle. the rectangular paddle
clearly performs better
6%
(see Figure 7b). Results are
5%

4%
mixed at the 90 RPM press
speed (see Figure 7c).
Weight Srel %

3%

2% Fill cam optimization.


1% Most presses offer a vari-
0% ety of fill cams to cover
10 20 30 40 50 60
a range of filling depths.
30 RPM 45 RPM 60 RPM 75 RPM 90 RPM The purpose of the fill
Feeder speed (RPM), round paddles cam is to overfill each
die and then push some
material back into the
The following example shows the impact of feeder feeder to ensure optimal die filling as the lower
speed on tablet weight variation, using a rectangular punch moves through the dosing cam. For exam-
feeder paddle (see Figure 5) and a round feeder paddle ple, a standard EURO or TSM B turret will offer
(see Figure 6), at different press speeds. This infor- a fill depth range of 018 mm. This filling depth
mation can be used to establish the optimal feeder is achieved by a range of filling cams, as follows:
speed at each press speed, to be stored in the product
recipe. In this example, it can be concluded that the 6 mm Fill Cam Fill Cam Range 06 mm
higher feeder speeds (> 40 RPM) and the rectangular 10 mm Fill Cam Fill Cam Range 010 mm
feeder paddle generally produce better results (i.e., 14 mm Fill Cam Fill Cam Range 414 mm
less weight variation), and the data can be used to 18 mm Fill Cam Fill Cam Range 818 mm.
32 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
Because there is over- Figure 7: Weight variation shown as relative standard deviation (Srel%) of tablet weight vs.
lap in the range of differ- feeder speed and feeder paddle configuration for rectangular paddles (blue lines) and round
paddles (red lines) at (a) 30 RPM, (b) 60 RPM, and (c) 90 RPM press speeds.
ent fill cams, the optimal
(a) 30 RPM press speed
fill cam must be selected
6%
empirically. For a prod- 5%

4%
uct that requires a final

Weight Srel %
3%
filling depth of 8 mm, 2%

the press can operate 1%

0%
with either the 10-mm 10 20 30 40 50 60

fill cam or the 14-mm (b) 60 RPM press speed


6%
f ill cam. Although it
5%
may seem logical that a 4%
Weight Srel %

deeper fill cam will be 3%

2%
better, that is not always
1%
the case. In summary, 0%
10 20 30 40 50 60
products that have fill-
(c) 90 RPM press speed
ing depths in the over-
lap between multiple fill 6%
5%
cams can be optimized
Weight Srel %

4%
only by testing each fill 3%
2%
cam across the desired 1%
0%
speed range.
10 20 30 40 50 60
Figure 8 plots the rela-
Rectangle Paddles Round Paddles
t ive s t a nd a rd de v ia-
Feeder speed (RPM)
tion of tablet weight at
different press speeds
Figure 8: Weight variation shown as relative standard deviation (Srel %) of tablet weight at
using two different fill
different press speeds using two different fill cams (10 mm, blue line and 14 mm, red line) at the
cams (10 mm and 14 same dosing setting and tablet weight.
mm) at the same dos- 6%

i ng s e t t i ng a nd t ab - 5%

4%
let weight. The results
Weight Srel %

3%
show compa rable re-
2%
sults through 60 RPM,
1%
but the deeper fill cam 0%
30 45 60 75 90
(14 mm) clearly extends
10 mm Fill Cam 14 mm Fill Cam
the process control (i.e.,
Press speed (RPM)
Contin. on page 45

Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 33


Continuous Manufacturing

Filling the Analysis Gap


in the Move to
Continuous Processing
Jamie Clayton

T
Effective analysis is key for he benefits of continuous manufacturing justify signifi-
the successful continuous cant investment, as evidenced by collaborations such as
manufacturing of solid- the MIT/Novartis Center for Continuous Manufacturing
dosage pharmaceuticals. (1). Batch production dominates within the pharmaceuti-
cal industry but many expect continuous processing to contribute a
substantial share of manufacturing capacity.
The move to continuous manufacturing requires changes in ana-
lytical practices in order to support new manufacturing models. Dy-
namic powder testing can contribute to the development of efficient
continuous processes.

The benefits of continuous manufacturing


Although innovative in many areas, the pharmaceutical industry
has historically focussed less on processing than other research
and development areas. Patent protection previously ensured that
R&D costs could be properly recouped, but as costs rise and time to
market increases, profitability cannot be guaranteed. When patents
expire, profitability relies on efficient production. Furthermore, a
regulatory focus on risk suggests a need for greater understanding of
processes and improved quality control. A shift from batch processes,
which are heavily dependent on manual intervention, to automated,
continuous operation is highly attractive.
In batch production, sequential steps are undertaken, with analysis
DAJ/GETTY IMAGES

performed in between. Batch-to-batch variability and products out


Jamie Clayton is
operations director at of specification (OOS) are common problems. The necessary rework
Freeman Technology.
and waste levels are unacceptably high.
34 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
THINI Download
Download
Article
Article
UPGRADE
Optimize your capability
Taking your processing capabilities to the next level is a strategy for
success; one that often demands upgrading equipment to meet new
production goals and future requirements. For immediate access to
the best technologies at the right price while maximizing returns from
surplus idle equipment, Federal Equipment Company is ready to help
you optimize operations while recovering equipment value through
accurate appraisals, strategic liquidations and full logistical support.

When you think equipment,


think Federal Equipment

1.877.536.1538 > www.fedequip.com


Continuous Manufacturing
Continuous processing is widely used in chemi- of a problem. With continuous manufacture, there
cal and food industries and offers the following is a question of how to define a batch. A batch be-
important advantages: comes associated with an operating period, which
tReduce costs, labor, and waste begins when start-up completes and ends at a de-
tOptimize asset utilization fined point. Any OOS products are therefore as-
t Simplify scale-up sociated with a time period rather than a discrete
tImprove containment. batch number, which can make problems difficult
These are environmental and economic im- to isolate.
provements, but there are also technical benefits. Optimized processing relies on understanding
A batch step has a beginning and an end; be- and controlling the materials and process variables
tween these points the product continuously that define clinical efficacy. The pharmaceutical
changes (e.g., a blending process starts with the industry has traditionally focused on developing
unmixed constituents and proceeds to a homo- and adhering to repeatable processes. This ap-
geneous state). A well-controlled continuous pro- proach relies on consistent feed and provides little
cess should operate at steady state for the major- flexibility to respond to variation. This is a critical
ity of the time. A steady-state operation requires limitation, as feed variability is a major source of
effective monitoring, as exemplified by widely failure. The transition from batch processing to-
used techniques such as in-line particle size and ward knowledge-based continuous manufacture,
near-infrared (NIR) analysis. Steady-state opera- however, has its challenges.
tion means continuous processing is associated
with consistent output, which equates to consis- A toolkit for more efficient manufacture
tent product quality. Scale-up is also simplified as Operations such as milling, roller compaction,
smaller units can be run for longer, avoiding the and tableting can be considered semi-continuous,
implications of changes in geometry and volume. as they are constantly fed during a batch cam-
Batch production does, however, have benefits. paign. The challenge involves engineering the
One benefit is flexibility; a suite of batch equip- equipment for reliable, prolonged operation and
ment can easily be reconfigured for different prod- successfully integrating the necessary compo-
ucts. Batch production also simplifies containment nents into an optimized continuous process. Au-

Center for Structured Organic Particulate Systems (C-SOPS)

For 10 years, researchers at the Center for Structured Organic Particulate Systems Test Bed 1 focuses on the simultaneous development of formulations, continuous
(C-SOPS) have worked to transform pharmaceutical manufacturing into a science- manufacturing, and analytical control methodologies for solid oral products. Test
driven discipline, in the areas of materials formulation and characterization, Bed 2 is designed to create an integrated continuous manufacturing platform to
design and scale up of material structuring, structural characterization and produce film-based drug products with controlled-release properties. Test Bed 3
modeling, and integrated systems science. C-SOPS also operates three test beds is based on drop-on-demand manufacturing, and uses liquid-phase processing to
to develop new continuous manufacturing processes. avoid challenges normally associated with conventional powder-based processes.
A National Science Foundation Engineering Research Center, C-SOPS is a Information about consortium members, research projects, and test bed
consortium of academic institutions and more than 40 industry partners from programs can be found at http://ercforsops.org.
both bio/pharmaceutical companies and industry equipment suppliers. The Editors of Pharmaceutical Technology

36 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
tomation is important but so are analytical tools Figure 1: Measuring flow energy to quantifying changes in flow
that provide the knowledge required to optimize properties following consolidation.

multi-component systems. The Engineering Re-


search Center for Structured Organic Particulate
Systems (C-SOPS) is a group at the forefront of

FLOW ENERGY / DENSITY


research in this area. The group applies modeling,
Flowability Change
>1000%
in-line analysis, and techniques such as powder
rheology to integrate sequential blending, dry
Density Change
granulation, lubrication, and tableting. A key 40% (max)

focus is to develop solutions to avoid three com- INCREASED TAPPING

mon tableting issues: segregation, agglomeration,


and compaction (2). of powders; however, the need for accurate, process-
Whether improving batch processes, or design- relevant data exposes limitations and highlights the
ing, monitoring, and controlling a continuous merits of techniques such as dynamic testing.
process, analytical tools are needed that deliver In dynamic testing, axial and rotational forces
relevant data and expand understanding of how acting on a blade are measured as it rotates through
processes work, reinforcing FDAs process analyti- a powder sample to determine values of flow en-
cal technology (PAT) initiative. PAT is defined as ergy that quantify how a powder f lows under
a system for designing, analyzing, and controlling conditions that reflect processing environments.
manufacturing through timely measurement of Powders can be characterized in consolidated, con-
critical quality and performance attributes of raw ditioned, aerated, or even fluidized states to mea-
and in-process materials and processes, with the sure the response to stress and air content. The
goal of ensuring final product quality (3). Real- impact of moisture, flow additives, compaction,
time analysis is therefore important but so are tech- attrition, and segregation can be evaluated.
niques that, for example, provide robust analysis of Figure 1 contrasts the change in bulk density in-
feeds prior to introduction to the plant. Identifying duced by tapping with the corresponding change
techniques that provide the information required to in flow energy. Flow energy increases by an order
achieve process efficiency is fundamental. of magnitude greater than density suggesting that
flow energy measurements are significantly more
Focus on powder testing sensitive in quantifying the impact of the change.
FIGURES ARE COURTESY OF THE AUTHOR.

Tablets are the most common drug-delivery vehicle, Furthermore, this indicates how density changes
and most drugs are handled in solid form at some could be misleading when quantifying how con-
point, demonstrating the need for suitable powder solidation impacts a process.
testing tools. Numerous methods for characteriz- This experiment emphasizes the importance
ing powders exist, including angle of repose, flow of selecting a suitable analytical technique for a
through an orifice, and tapped density. These sim- given application. It is increasingly acknowledged
ple techniques provide some insight into the nature that no single powder test suits every application
Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 37
Continuous Manufacturing
Figure 2: Dynamic testing can provide differentiation of materials that shear tests classify as identical.

2 vanillin 1800
2 ethylvanillin
5 1600

1400
4
1200

Total energy, mJ
Shear stress. kPa

1000
3

800

2
600

400
1 2 vanillin
200 2 ethylvanillin

0
0
Test number 0 2 4 6 8 10
0 1 2 3 4 5 6 7 8 9
Tip speed, mm/s -100 -100 -100 -100 -100 -100 -100 -100 -70 -40 -10
Applied normal stress, kPa

and that tests should represent the conditions to process design and enable effective monitoring
which the material is exposed. and control. It is essential to consider what in-
Shear testing, for example, is a well-estab- formation is required and how to obtain it. Ap-
lished technique developed to support hopper plying this approach to powder characterization
design protocols advanced by Jenike (4, 5). It is highlights limitations with traditional techniques.
still widely applied, with modern instrumenta- Innovative techniques such as dynamic testing
tion delivering improved reproducibility, and is and, in particular, instruments that combine dy-
a valuable tool for characterizing a powder in a namic testing with methods such as shear and
static, consolidated state, but there are limitations. bulk property analysis, present an efficient and
Figure 2 shows shear and flow energy data for versatile choice for those demanding new levels
two excipients: vanillin and ethylvanillin. Shear of efficiency.
testing suggests these materials are identical,
while dynamic testing identifies clear differ- References
1. Novartis-MIT Center for Continuous Manufacturing website,
ences. In this case, the f low energy correlated https://novartis-mit.mit.edu/, accessed March 1, 2016.
with in-process behavior, illustrating how mate- 2. Center for Structured Organic Particulate Systems, Test Bed 1,
Continuous Powder Manufacturing, http://csops.rutgers.edu/
rials classified as identical by shear testing may research/test-beds/test-bed-1, accessed March 1, 2016.
3. FDA, Progress Report on Process Analytical Technology,
actually process differently. This highlights the www.fda.gov/drugs/developmentapprovalprocess/manufac-
importance of employing methods that simulate turing/questionsandanswersoncurrentgoodmanufacturing-
practicescgmpfordrugs/ucm072006.htm, accessed March 1,
process conditions. 2016.
4. A.W. Roberts, Basic Principles of Bulk Solids Storage, Flow and
Handling (The Institute for Bulk Materials Handling Re-
Looking ahead search, Callaghan, NSW, Australia, 1993).
5. The Institution of Chemical Engineers/European Federation
The rise of continuous manufacture, and the of Chemical Engineering, Standard Shear Testing Technique
drive for greater efficiency, increases the need for Particulate Solids Using the Jenike Shear Cell, A Report of
the EFCE Working Party on the Mechanics of Particulate Sol-
for analytical techniques that support intelligent ids (1989). PT

38 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
Download
Case Study

I need a CDMO
thats a true PARTNER.
With Rottendorf Pharma as your CDMO, you can be certain you are working with
a financially stable company committed to you in a long-term relationship. We
will know your goals and bring proven ideas to the table on how to achieve them.
Need a true partner for your solid oral doses? Rottendorf owns it.

www.Rottendorf.com/partner

M A N U FA C T U R I N G PA C K A G I N G F O R M U L AT I O N D E V E L O P M E N T A N A LY T I C A L M E T H O D S
Elemental Impurities

Meeting USP Guidelines


for Elemental Impurity
Analysis with X-ray
Fluorescence Spectrometry
Andrew Fussell

T
This article discusses the here are strict limits and clear regulatory guidelines for the
USP guidelines for monitoring of elemental impurities in drug products. An
monitoring elemental
important source of these impurities is the catalysts, which
impurities in drug products.
are necessary during drug production. These catalysts in-
clude palladium, platinum, rhodium, iridium, and rutheniumall
elements that, at high concentrations, can be harmful to patients.
Previously, the United States Pharmacopeia (USP) chapter <231> (1)
set the standard for the control of elemental impurities; however,
the qualitative approach lacked selectivity and sensitivity and could
fail to detect certain elements, such as mercury, at toxicologically
relevant levels.
For some time, it has been widely recognized within the phar-
maceutical industry that the methodology set out in USP <231> is
outdated, compared to more modern quantitative techniques. In
response, the US Pharmacopeial Convention (USP) has developed
new regulations, chapters <232> and <233> (2, 3), through a pro-
cess of consultation with the industry, chemists, and toxicologists.
USP <232> and <233> contain changes to the concentration limits
for elemental impurities, as well as introducing flexibility in the
TETRA IMAGES/GETTY IMAGES

choice of testing methods. In an effort to harmonize the regula-


tions, the International Council for Harmonization (ICH) worked
together with representatives from the European Pharmacopoeia
Dr. Andrew Fussell is the
pharmaceutical segment (Ph. Eur.), the Japanese Pharmacopeia, and USP to create the ICH
manager at PANalytical.
Q3D guideline for elemental impurities (4). ICH Q3D has been ad-
40 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
opted verbatim by the European Medicines Agency Table I: Twenty elements spiked into pure cellulose, lactose, or
(EMA). The ICH Q3D guideline does not stipu- calcium carbonate matrix. International Council for Harmonization
(ICH) oral delivery risk assessment elements shown in bold.
late analytical procedures but instead states that Concentration PAN Concentration
PAN standards
range (g/g) standards range (g/g)
pharmacopeial procedures or suitable alternative
As 0-100 Se 0-200
procedures should be used. These regulations (USP
Cd 0-100 TI 0-200
and ICH Q3D) will apply both to products newly Pb 0-100 Cu 0-1000
released onto the market and legacy products, which Hg 0-100 Zn 0-1000
will need to be retested. Co 0-200 Mo 0-200

In a significant move, USP <233> allows manu- V 0-200 Ru 0-200

facturers choice in the analytical method used for Ni 0-1000 Rh 0-200

the evaluation of levels of elemental impurities so Cr 0-500 Pd 0-200

Mn 0-1000 Ir 0-200
long as the technique has been validated in line
Fe 0-1000 Pt 0-200
with the requirements. One option emerging as
a favored alternative is X-ray fluorescence spec- ing. It can be used for the elemental and chemical
trometry (XRF). analysis of solid, powdered, and liquid samples,
making it particularly valuable to pharmaceuti-
X-ray analysis in pharmaceuticals cals. One of the main advantages of XRF is that it
Reliable, accurate analytical processes are the basis requires minimal if any sample preparation prior
for most activities in the pharmaceutical industry to analysis. In addition, the instrumentation is well
and X-ray-based analytical techniques underpin adapted to automation.
many of these procedures. X-ray diffraction (XRD) This is in sharp contrast to inductively coupled
is an established technique in the industry and is plasma atomic emission spectroscopy (ICPAES)
widely used for qualitative and quantitative analy- and inductively coupled plasma mass spectrometry
sis of solid phases (5). XRD has long been the ac- (ICPMS), which feature in the two sample meth-
cepted technique for establishing the crystalline ods outlined in USP <233>. ICPAES uses ICP to
drug fingerprint needed for drug approval, pat- excite the atoms in a sample to emit electromag-
ent descriptions, and for the identification of dif- netic radiationthe wavelength indicates the pres-
ferent drug batches (6). The use of XRD has high- ence of an element while the intensity indicates the
lighted the general benefits of X-ray analysis to the concentration. In ICPMS, ICP is used to ionize
pharmaceutical industry, including the following: a sample, while mass spectrometry separates and
t Speed of analysis quantifies the elements present. Both of these tech-
t Simple (or no) sample preparation niques are already used in parts of the industry;
tNon-destructive measurement. however, the sample preparation is intensive, and
In parallel, XRF is considered a proven tech- the dilutions required can lead to errors in analysis.
nique for material analysis in a broad range of in- XRF analysis can be divided into wavelength
dustries and applications from measuring sulfur in dispersive (WD) and energy dispersive (ED) tech-
oil to analyzing coating thickness in metal finish- niques. The difference between these two tech-
Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 41
Elemental Impurities

Table II: International Council for Harmonization (ICH) and United States Pharmacopeia (USP) limits and validation sample set up. PDE is
permissible daily exposure.
USP conc. limit Calc. USP max.
ICH threshold Lower spike conc. Upper spike conc.
Element ICH PDE (g/g) for oral drug daily dose (g/
(30%) (g/g) (g/g)
products (g/g) day)
As 15 4.5 3 5 1 9

Cd 5 1.5 2 2.5 1 9

Pb 5 1.5 2 2.5 1 9

Co 50 15 10 5 5 20

V 100 30 10 10 5 20

Ni 200 60 20 10 5 60

Cu 3000 900 300 10 15 150

Cr 11000 3300 1100 10 5 75

Mo 3000 900 300 10 5 120

Ru 100 30 10 10 5 15

Rh 100 30 10 10 5 15

Pd 100 30 10 10 5 15

Ir 100 30 10 10 5 15

Pt 100 30 10 10 5 15

inner-shell electrons of the elements in the sample.


Table III: Limits of detectionEpsilon 3X; PDE is permissible
daily exposure; LLD is lower limit of detection. The interaction of the high energy X-rays with the
ICH PDE ICH threshold LLD (g/g) Measurement
Element
(g/g) (30%) (3m of blank) time (min) electrons results in the emission of X-rays of char-
As 15 4.5 0.1 acteristic energies for the elements in the sample.
30
Pb 5 1.5 0.1 The WD technique uses an analyzing crystal to
Cd 5 1.5 0.4 30 separate the characteristic X-rays for the different
Co 50 15 0.1
30 elements, which are then detected by an X-ray de-
Cr 11000 3300 0.2
tector. The ED techniques do not require an ana-
Ni 200 60 0.1
lyzing crystal, but instead, the X-ray detector itself
Cu 3000 900 0.2 15

Mo 3000 900 0.4


determines both the energy and the intensity of
V 100 30 0.2 15 the characteristic X-rays (7).
Ru 100 30 0.4 In this study, EDXRF systems were used. EDXRF
Rh 100 30 0.9 30 spectrometers discriminate each specific X-radia-
Pd 100 30 0.9 tion line based on the energy of the produced pho-
Ir 100 30 0.3
5 ton. Compared to other techniques, EDXRF spec-
Pt 100 30 0.3
trometers offer a number of advantages in that they
niques is due to the different detection methods. tend to be smaller, simpler in design, faster, have
Both techniques typically use an X-ray tube to fewer engineered parts, and are typically cheaper.
generate high energy X-rays that interact with the In addition, the advances in XRF technology in
42 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
recent years make this method particularly attrac- Table IV: Limits of detectionEpsilon 5; PDE is permissible daily
tive to the pharmaceutical industry, particularly exposure; LLD is lower limit of detection
ICH PDE ICH threshold LLD (g/g) Measurement
as it is now a pharmacopeial method with USP Element
(g/g) (30%) (3m of blank) time (min)

<735> X-ray fluorescence spectrometry and Ph. As 15 4.5 0.8 10

Eur. chapter 2.2.37. Cd 5 1.5 0.3 10

Pb 5 1.5 0.3 10

Co 50 15 1.7
In practice
Ni 200 60 0.4 5
It is recognized that the greatest risks in drug sub-
Cu 3000 900 7.2
stances is from intentionally added metals, namely V 100 30 0.4
5
metal catalysts. To combat this risk, the industry Cr 11000 3300 0.3
has adopted a risk-based approach to assessing the Mo 3000 900 0.2 5

potential presence of elemental impurities in drug Ru 100 30 0.2


5
products. During the manufacture of drugs, risk Rh 100 30 0.1
Pd 100 30 0.4 5
assessments have to be conducted that are in ac-
Ir 100 30 0.3
cordance with USP <232> or ICH Q3D, which lists 5
Pt 100 30 0.4
the permissible daily exposure (PDE) for potential
impurity elements in pharmaceutical materials. Table V: Calibration results for the Epsilon 5. RMS is root mean squared.
To demonstrate how XRF fulfills the guideline Element
Conc. range
R value RMS error (g/g)
(g/g)
criteria, pharmaceutical calibration standards for As 0-100 0.9998 0.6
elemental impurity analysis were developed by Cd 0-100 0.9999 0.3
spiking 20 elements into pure cellulose, lactose, Pb 0-100 0.9989 1.6

or calcium carbonate matrix, as shown in Table I. Co 0-200 0.9986 3.7

The calibration samples were prepared in triplicate V 0-200 0.9999 1.0

Ni 0-1000 0.9998 6.7


using 1 g of loose powder, using a 6 m polypro-
Cu 0-1000 0.9998 7.0
pylene foil. The results presented in this work are
Cr 0-500 0.9999 2.9
for the cellulose matrix.
Mn 0-1000 0.9996 10.8
The validation samples were also prepared in Mo 0-200 0.9994 2.5
triplicate by mixing pure paracetamol (acetamino- Ru 0-200 1.0000 0.6
phen) with known concentrations of test elements Rh 0-200 1.0000 0.5

from the calibration materials. Each test element Pd 0-200 0.9997 1.7

was validated at two different concentrations as Ir 0-200 0.9999 1.1

Pt 0-200 0.9998 1.5


required by USP <233>. In most cases, the vali-
dation samples were above and below the target
concentration. The powders were mixed for one was prepared and filled with 1 g of sample. Next,
minute in plastic vials with ZrO2 milling balls. The the sample was inserted into the instrument for
total sample preparation took less than five min- measurement. Table II shows the sample set up used.
utes. For analysis, a loose powder sample holder The first system used was the Epsilon 3X, which
Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 43
Elemental Impurities
Table VI: Validation spike results for Epsilon 5. USP is United States Pharmacopeia.
Expected spike conc. Measurement conc. Within USP<233> Measurement time
Element Percentage recovery
(g/g) (g/g) limit (70-150%) (min)
As 1 1.19 0.03 119 YES 5

9 8.87 0.22 99 YES 10

Cd 1 0.77 0.14 77 YES 5

9 8.30 0.12 92 YES 10

Pb 3 2.78 0.21 93 YES 5

9 10.33 0.15 115 YES 10

Co 5 6.90 1.32 138 YES 5

20 16.29 0.01 81 YES 3

V 5 6.52 0.13 130 YES 5

20 23.03 0.31 115 YES 2.5

Ni 5 5.42 1.29 108 YES 5

60 57.23 2.34 95 YES 3

Cu 15 16.30 1.58 109 YES 5

150 144.36 6.87 96 YES 3

Pd 5 4.98 0.11 100 YES 3

15 14.48 0.04 97 YES 3

Figure 1: Calibration lines for As (left), Cd (middle), and Pd (right) show good linearity.

0.10

0.04
0.10

0.03

0.05
0.05 0.02

0.01

0.00 0.00 0.00


0 50 100 0 50 100 0 50 100 150 200
Concentration (g/g) Concentration (g/g) Concentration (g/g)

is a benchtop EDXRF spectrometer with a 10-posi- The samples were also analyzed with the Epsi-
tion sample changer. The analysis was performed in lon 5, a floor standing EDXRF spectrometer with
air atmosphere not requiring any external gasses. A a 133-position sample changer. For this instrument,
total one off calibration time was 15 hours for all 20 the measurements were performed in helium at-
elements. The measurement time used to obtain the mosphere. The total one off calibration time was
detection limits (Table III) was between 530 minutes significantly reduced when compared with the Ep-
per element, while the validation spike sample mea- silon 3X at 6 hours for all 20 elements. The limit
surement was between 2.530 minutes per element. of detection measurement time and the validation
Measurement time can be shortened depending on spiked sample measurement time were also re-
the number of elements, sample size, and accuracy duced to 510 minutes and 2.510 minutes per ele-
and precision requirements needed. ment, respectively. Again, this time can be reduced
44 Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 P h a r mTe c h . c o m
depending on the number of elements, sample size, as reduced potential for error through minimized
and the accuracy and precision requirements. The sample preparation, the non-destructive nature
limits of detection for the Epsilon 3X and Epsilon of the analysis, reduced chemical waste, and the
5 are shown in Tables III and IV. lower total cost of ownership and operation. Many
Good linearity was achieved for all the elements companies are picking up the opportunity to use
tested (as noted in Table V), Examples of the cali- alternative techniques and taking advantage of the
bration curves for As, Cd, and Pd are shown in methods now available. It is important for these
Figure 1. Repeatability was ascertained by analyz- techniques to be fully explored, as it will allow
ing the standards measured 20 times consecutively the pharmaceutical industry to find the technique
as unknowns. Further, the validation spike results and methods that best suit their requirements.
shown in Table VI show a reasonable recovery per-
centage for a number of elements mixed as pow- References
1. USP, Chapter <231>, USP 35NF 30, (USP, 2012).
ders with paracetamol. 2. USP, Chapter <232>, USP 35NF 30, (USP, 2012).
3. USP, Chapter <233>, USP 35NF 30, (USP, 2012).
4. ICH, Q3D Guideline for Elemental Impurities (ICH, 2014),
Conclusion www.ich.org/fileadmin/Public_Web_Site/ICH_Products/
Guidelines/Quality/Q3D/Q3D_Step_4.pdf, accessed Feb. 22
By allowing the use of alternative techniques, such 2016.
as XRF, USP is now supporting pharmaceutical 5. Vyas K., X-ray Diffraction in Pharma Industry (2013), www.
icdd.com/ppxrd/12/abstracts/P26.pdf, accessed Feb. 22, 2016.
manufacturers who want to make their own ana- 6. Randall C., Rocco W., Ricou P. , XRD in Pharmaceutical Analy-
sis: A Versatile Tool for Problem-Solving (2010), www.american-
lytical choices. EDXRF was shown to be a pow- pharmaceuticalreview.com/Featured-Articles/115052-XRD-in-
erful technique capable of reaching the required Pharmaceutical-Analysis-A-Versatile-Tool-for-Problem-Solv-
ing/, accessed Feb. 22, 2016.
USP <232> and ICH Q3D PDE limits for oral 7. Brouwer P., Theory of XRFGetting acquainted with the prin-
drugs. XRF also provides a number of advantages ciples (booklet) (2013), www.panalytical.com/Technology-
background/Theory-of-XRF-getting-acquainted-with-the-
over the USP <233> wet chemical techniques, such principles-booklet.htm, accessed Feb. 22, 2016). PT

Tablet Compression contin. from page 33


maintains low weight variation) through the 75 necessary to expand the process optimization tem-
RPM press speed. plate to incorporate representative granulations for
nominal and more difficult batches.
Conclusion Most modern tablet presses incorporate a prod-
Applying a methodical approach to tablet-com- uct recipe capability that will permit optimized
pression process optimization and evaluating the press settings to be stored and automatically re-
process control under a range of different condi- trieved to eliminate redundant optimization efforts
tions will permit a full understanding of the pro- and expensive losses at start-up. In many cases,
cess and will facilitate the opportunity to achieve there are opportunities to implement process opti-
optimal tablet quality and production output. This mization measures on existing products and obtain
template assumes limited batch-to-batch variabil- benefits from the resulting improvements in tablet
ity. If batch-to-batch variability is high, it may be quality and production efficiencies. PT
Pharmaceutical Technology SOLID DOSAGE DRUG DEVELOPMENT AND MANUFACTURING 2016 45

Вам также может понравиться