You are on page 1of 24

C H A P T E R

8
Particle, Powder, and Compact
Characterization
Gregory E. Amidon, Pamela J. Secreast and Deanna Mudie

8.1 INTRODUCTION There are advantages and disadvantages to the mate-


rial sparing paradigm. Using smaller amounts of mate-
rial will result in a lower cost, but may increase the risk
As timelines become tighter, internal and external
of working with a non-representative sample, which may
pressures abound for the pharmaceutical industry.
result in errors. In order to reduce resource requirements,
The cost of drug development continues to rise at an
we have proposed strategies to decrease method devel-
alarming rate (with an average cost to market of $800
opment time and introduced general guidelines.
million per drug), making it imperative to develop
drugs in more effective ways to reduce cost and time.
Historically, early drug product development has 8.2 PARTICLE SIZE
focused on using design of experiments to manufac-
ture numerous “kilogram size batches” to optimize
CHARACTERIZATION
the formulation, and gain a complete understanding
of the manufacturing process. This method may pro- Particle characterization is an important compo-
duce a high-quality product, but requires the use of nent of data driven formulation development on a
large quantities of expensive active pharmaceutical material-sparing scale. Particle size, size distribution,
ingredient (API), and results in lengthy timelines. shape, and texture can all have an impact on pharma-
Since most compounds do not make it through the ceutical processing and performance, hence consider-
development stages, a different approach is needed ation must be given to the impact of these parameters
for product development. Recent efforts have focused on the robustness of processing. This is true for API,
on using material-sparing strategies to develop early excipients, and formulations (blends and granula-
solid dosage forms, such as powder-in-capsule and tions). When characterizing API, excipients or formu-
immediate release tablets. Formulations are developed lations, a representative sample must be obtained. The
by characterizating particle, powder, and compacts of sample needs to be correctly and reproducibly pre-
API, excipients, and formulations at small scale (50– pared, and instrumental parameters must be correctly
200 grams). Such characterization, coupled with some used for the analysis.1 Each particle sizing method,
predictive tools, allows scientists to understand the replete with its own assumptions, will result in a
important physical, chemical, and mechanical proper- unique measure of particle size and distribution, since
ties of materials to design robust formulations to for- a real sample exhibits a range of shapes and sizes
mulate quickly, at a relatively low cost, and to achieve whose complexity is compounded when analyzing
acceptable exposure in clinical studies. Some of the multi-component systems such as formulations. Since
characterization methods discussed in this chapter are no two methods of particle sizing will result in the
summarized in Table 8.1. same “numbers,” it becomes important to distinguish

Developing Solid Oral Dosage Forms: Pharmaceutical Theory and Practice 163 © 2009, Elsevier Inc.
164 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

TABLE 8.1 Material-sparing characterization methods later in this chapter) a solid particle is considered to
approximate a sphere. Since this measurement is based
Method Measured parameters on a hypothetical, not actual, sphere and is only an
approximation of the true particle shape, it is referred to
Particle characterization as the equivalent spherical diameter of the particle.3
Light microscopy Size, shape, roughness, size range For other analyses of irregularly shaped particles
Polarized light microscopy Crystallinity (such as microscopic evaluation), information about
Scanning electron microscopy Size, shape, roughness, size range both size and shape is needed. There are several differ-
Sieving Size, size distribution ent particle diameters that can be used in the evaluation
Light diffraction particle size Quantitative size, distribution, of particle size; the most common of these are illus-
span trated in Figure 8.1.4 While length and width are diam-
Powder characterization eters more often used in microscopic evaluation, the
Helium pycnometry True density equivalent spherical diameter is widely used in laser
Bulk/tapped density Bulk and tapped density, light diffraction analyses. Another descriptor of parti-
compressibility index cle diameter used in early formulation development is
Shear cell Powder flow parameters, flow the sieve diameter, which is the width of the minimum
function coefficient, unconfined square aperture through which a particle will pass.
yield strength, cohesion, effective Descriptors of particle shape include acicular,
angle of internal friction
columnar, flake, plate, lath and equant, and should be
Compact characterization included in the characterization of irregularly shaped
Tablet compaction Compaction pressure, solid particles. Degree of particle association (e.g., aggre-
fraction
gate, agglomerate), and surface characteristics (e.g.,
Indentation test Deformation pressure, elastic cracked, smooth, porous), should also be noted.4
deformation
Since most APIs, excipients, and formulations are
Tensile test Tensile strength, compromised
found in a range of sizes, size distribution of the mate-
tensile strength
rials must also be determined. When a bell-shaped
Tablet characterization
curve resembling a normal distribution can describe
Tabletability Tensile strength—solid fraction
particle size distribution, it can be described by the
relationship
mean particle size and standard deviation. Again,
Compactibility Tensile strength—compression
pressure relationship
however, many pharmaceutical powders do not fall
into a bell-shaped distribution. Some are skewed
Compressibility Solid fraction—compression
pressure relationship toward the upper end of the particle size range. These
Manufacturability Tablet crushing force—
powders are better described using a log-normal dis-
compression force relationship tribution that, when the data are plotted using a log
scale for the size axis, resembles a normal distribu-
tion curve. When using a log-normal representation
the geometric mean standard deviation, and geomet-
between different methods, and to use the same type ric median particle size, are used to describe the dis-
of analysis when comparing lots of API, excipients or tribution. Other pharmaceutical powders cannot be
formulations, examining differences in performance characterized by a single distribution, and may have
that process changes make, etc. While many types of bimodal or multimodal distributions. Cumulative fre-
analyses for particle characterization are available and quency distribution curves may also be generated by
possible, this discussion will focus on those methods adding percent frequency values to produce a cumu-
used in the material-sparing paradigm. Table 8.2 out- lative percent frequency distribution. Sieving data is
lines the various techniques used for particle character- often presented in this manner.
ization in a material-sparing formulation development
process. For those interested in more detailed informa-
tion on particles size measurements, standard textbooks
are recommended.2
8.2.1 Light Microscopy
Particle size measurements vary in difficulty depend- Light microscopy is perhaps the ultimate material-
ing on the shape of the particle. Spherical particles, for sparing characterization technique, and forms the basis
example, can be fully defined by their diameter. In real- of the material-sparing approach as it pertains to parti-
ity, most particles are far from spherical; however, for cle characterization. With only a few milligrams of mate-
certain analyses (such as laser light diffraction, discussed rial, appearance, shape, extent of particle association,

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.2 PARTICLE SIZE CHARACTERIZATION 165

TABLE 8.2 Summary of material-sparing particle characterization techniques

Min, Max,
Method micron microns Distribution Shape Texture Grams Crystallinity

Microscopy
Light 1 1000 Yes Yes Yes 0.1 No
Polarized 1 1000 Yes Yes Yes 0.1 Yes
SEM 0.02 1000 Maybe Yes Yes 0.1 No
Dynamic image 1 10000 Yes Yes No 2–5 No
analysis
Light scattering
Laser—wet 0.02 2000 Yes No No 1–2 No
Laser—dry 0.02 2000 Yes No No 1–2 No
Other
Sieving 25–50 2000 Yes No No 3–5 No

For a compound light microscope, the type of


n gth microscope generally used, the practical size range is
Le 1 micron to 1 millimeter. This type of microscope is
used when fine detail must be resolved. The clarity or
Feret’s diameter
resolving power of the image depends on the sharp-
ness of the image produced by the objective lens.
Resolving power is defined as the smallest distance
Martin’s diameter between two points such that the two points can be
Projected area diameter
distinguished.5 The sample is usually mounted in a
medium that has sufficient contrast to the sample, and
Maximum horizontal
in which the sample is insoluble. The particles should
all be in the same plane, be adequately separated from
Wi
dth

each other, and should represent the size and distribu-


tion of the entire sample population.
FIGURE 8.1 Particle shape parameters

and texture information may be quickly obtained.


With a polarizing light microscope, extent of crystal-
8.2.2 Scanning Electron Microscopy
linity may also be assessed. Particle size distributions Scanning electron microscopy (SEM) is another
(PSD) may be generated from microscopic evalua- technique where only milligram quantities of mate-
tion, although it is a tedious and time-consuming rial may be used to determine particle size, shape, and
process. Laser diffraction has become increasingly texture. In SEM a fine beam of electrons scan across
popular for PSD, and will be discussed later in the the prepared sample in a series of parallel tracks. The
chapter. More recently, dynamic image analyzers have electrons interact with the sample, and produce sev-
been developed which automate the analysis of par- eral different signals which can be detected and dis-
ticle size and shape by capturing images with a cam- played on the screen of a cathode ray tube.2 Particles
era, transmitting them to a computer system, and then less than 1 nm can be viewed, and since the depth of
analyzing them electronically. Average particle size, focus is so much greater than that of the light micro-
size distribution, and shape information may be gener- scope, information on surface texture can be gener-
ated using this technique. Image analysis may be used ated. SEM requires more time-consuming sample
for particles in the size range of 1 micron to 10 mm, but preparation than optical microscopy, and cannot
requires up to several grams of material depending on distinguish between crystalline and non-crystalline
the difficulty of method development, and thus far has materials. It is also more difficult to generate a particle
not been routinely used during the material-sparing size distribution using SEM since, while the informa-
formulation process. tion obtained is visual and descriptive, it is usually

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


166 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

not quantitative since only a few particles are seen in LD methods. Analysis time is short, robust methods
the viewing field at one time. However, when SEM is can be developed with a minimum of material, results
used with other techniques such as laser diffraction, it are reproducible, calibration is not required, and a
can provide valuable additional information on parti- wide range of measurement is possible. LD technology
cle texture, which may help to explain agglomeration is divided into two general methods. High angle light
or flow problems. scattering is appropriate for very small (submicron)
particles, and falls outside the scope of this discus-
sion. Low angle light scattering is used for pharma-
ceutical materials in the micron size range or greater.
8.2.3 Sieving
Determination of particle size using low angle tech-
Sieving is a simple method that is used for deter- nology is only effective if one has a model to interpret
mining the particle size distribution of a powder. It is it with. For particles much larger than the wavelength
often the preferred method of choice for formulators, of light (5 microns) any interaction with those parti-
since it is a straightforward analysis that can be done cles causes the light to be scattered with only a small
during the formulation development process (after change in angle. This is known as Fraunhauer diffrac-
mixing or granulation of a formulation, for example). tion, and produces light intensity patterns that occur
This technique is most often used for formulations at regular intervals and are proportional to the parti-
or excipients since larger quantities are needed (3–5 cle diameter that produces this scatter. Fraunhauer
grams is needed to perform an analysis using small- diffraction-based instruments are applicable for parti-
scale sonic sifting). API is not usually evaluated by cle size ranges of about 1–2000 microns, depending on
sieving due to the particle size limitations, as well as the lens used.
the more irregular particle shapes. Sieving is most Both wet and dry laser diffraction methods are
suitable for powders whose average particle size is possible, and are used in a material-sparing process.
greater than 25–50 microns.1 For dry dispersion LD, initial conditions such as pres-
For material-sparing sieving, a sonic sifter using sure and lens size are determined by microscopy, with
3-inch sieves is employed. Sonic sieving combines two a short series of trial runs to assess correct pressure
motions to separate the particles: a vertically oscillat- for dispersing particles without fracturing them and
ing air column to lift particles and carry them back to confirm lens size. Too high a pressure may break
against mesh openings at several thousand pulses per down some fragile particles. With small tray acces-
minute, and a repetitive tapping pulse to help reduce sories, these dry methods can use 500 mg or less for a
sieve blinding. Tapping also helps to deagglomer- complete analysis. Initial conditions for a wet method
ate samples with electrostatic, hygroscopic or other rely on a small set of standardized generic methods
adhesion problems.6 The raw data can be converted and microscopy. For wet methods it is important to
into a cumulative weight distribution, and the data choose a suspension medium where the sample has
presented as a percentage of the sample retained on low solubility (hexane is often used) that will also
each sieve (histogram) or a cumulative distribution disperse the sample adequately. Surfactants are occa-
as a function of sieve size. When data is plotted on a sionally used to facilitate dispersion and inhibit floc-
log–log plot (sieve size versus cumulative percent), culation of the sample, but it is important to prevent
unimodal versus polymodal distributions can also be particle dissolution in the surfactant solution. A fil-
determined. A detailed procedure for analytical siev- tered saturated solution can be used as the dispers-
ing may be found in the USP general method 786 , ing medium to effectively prevent particle dissolution
Method I (dry sieving).7 in the medium. Sonication may be needed to break
up aggregates, but fragility of the sample must be
assessed since sonication may fracture individual
particles and skew the results. Sample load is also
8.2.4 Light Diffraction
important as a higher percentage of the sample in the
Laser light diffraction (LD) is becoming a preferred dispersing solvent may cause aggregation.
method for the determination of particle size distribu- Whether a wet or dry method is used during material-
tion of pharmaceutical materials. This technique can sparing formulation development depends on several
be quite material sparing, using 1–3 grams of a mate- factors, the most important of which are the properties
rial for a complete evaluation. Even less material may of the material to be tested. Wet dispersion is frequently
be used if combined with a microscopic or SEM evalu- used for API characterization since it is a one-component
ation to determine particle size, morphology, and fra- system, easily screened for the appropriate non-soluble
gility of the sample. There are several advantages to dispersant. Multi-component systems, such as mixes

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.3 POWDER CHARACTERIZATION 167

or granulations, are more amenable to dry dispersion σg d90/d50


techniques. There is no need to test the solubility of each 1.0 1.0
1000

Maximum geometric mean volume particle diameter (d50), m


component in the dispersant, since the dispersant for 1.5 1.7

dry techniques is air. Dry dispersion techniques tend to 2.0 2.4


be used when there is plenty of material available, since
developing a method is straightforward with sufficient 2.5 3.2

trial runs. Other factors that determine which method is 3.0 4.1
utilized include operator experience, equipment avail-
ability, and personal preference. 100
3.5 4.9

4.0 5.8

Milling
8.2.5 Importance/Impact of Particle Size
Characterization
Understanding a pharmaceutical powder’s par-
ticle size, shape, and distribution is an important 10

component of material-sparing formulation devel-

Micronizing
opment. When working with the API, a few large or
small particles in a batch can alter the final tablet’s
content uniformity (potency, segregation), dissolu-
tion profile, and/or processing (e.g., flow, compres-
sion pressure profile, and granulating properties if it
1
is for dry granulation). Rohrs et al. have shown that, 0.1 1 10 100 1000
with only the predicted API dose and the geometric Dose, mg
standard deviation, an estimate of the API particle FIGURE 8.2 Content uniformity prediction as a function of par-
size can be obtained that will have a 99% probability ticle size, distribution, and dose
of passing USP Stage I content uniformity criteria.
This is displayed graphically in Figure 8.2. It is use-
ful for determining approximate figures for particle necessary to achieve 80% dissolved in a USP dissolu-
size requirements, including whether additional API tion apparatus in 30 minutes under sink conditions as
processing such as milling is necessary.8 API parti- a function of solubility and particle size distribution
cle size and distribution data information can also (sigma  geometric standard deviation of a log-nor-
help decide whether a direct compression formula- mal distribution) is shown in Figure 8.3.12 With this
tion or dry granulation approach is most suitable. information, selection of an appropriate particle size
Examination of the API can also reveal inter- and and distribution needed to achieve the desired disso-
intra-batch differences and/or trends. If, for exam- lution rate may be estimated.
ple, the particle size distribution has changed from Particle characterization of the in-process or final-
one batch of API to the next, this could significantly formulation is also critical. Flow characteristics of
impact the processability of the final formulation. formulations (which will be discussed later in the
Particle size and size distribution are also impor- chapter) are based, among other factors, on shape,
tant, from a dosage form performance point of view, size, and size distribution of particles. Large, spherical
in that they are critical parameters in assuring that the particles flow better than smaller, irregularly shaped
desired dissolution rate is achieved for oral dosage materials, for example.
forms. Several theoretical models for dissolution of
powders have been developed.9,10,11 Using a Noyes–
Whitney type expression as a starting point, Higuchi
and Hiestand,10 as well as Hintz and Johnson,11 8.3 POWDER CHARACTERIZATION
derived expressions for dissolution of spherical parti-
cles as a function of time. While details are beyond the 8.3.1 Density
scope of this chapter, it is possible to predict the dis-
solution rate of poly-dispersed particle size distribu- True Density
tions by summing up the predicted dissolution rate of The true density of a substance is the average mass
individual size fractions of the powder. For example, of the particles divided by the solid volume, exclusive
using this approach, the predicted particle diameter of all the voids that are not a fundamental part of the

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


168 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

100 Bulk Density


Particle diameter to achieve 80%

Bulk density is the mass per unit volume of a loose


dissolved in 30 min

powder bed. The unit volume includes the spaces


between the particles, and the envelope volumes of
10 the particles themselves. The method used to fill the
material into that volume can affect the degree to
Monodispersed
which the powder is compressed, and can thus influ-
Sigma  2.0
Sigma  3.0
ence the bulk density value.14,15 Bulk density can
1
be calculated using Equation 8.2, where M  mass
1 10 100 1000 in grams and Vo  untapped apparent volume in
Solubility in μg/mL milliliters.
FIGURE 8.3 Predicted particle diameter necessary to achieve
80% dissolved in a USP dissolution apparatus in 30 minutes under M
sink conditions as a function of solubility and particle size distri- Bulk Density(g/mL )  (8.2)
Vo
bution (sigma  geometric standard deviation of a log-normal
distribution)
The loose or “aerated” bulk density can be deter-
mined by allowing a defined amount of material to fill
molecular packing arrangement; therefore, it should a container with a known volume under the influence
be independent of the method of determination.13 of gravity.17 The amount to which the particles col-
There are three basic methods for determining true lapse, and fill voids between the particles, will depend
density: on a number of powder properties, including particle
● gas pycnometry or displacement; shape, particle size distribution, inter-particle friction,
● liquid displacement; and and cohesion.
● flotation in a liquid. Bulk density is typically measured by gently intro-
ducing a known sample mass into a graduated cyl-
Gas pycnometry is used in the material-sparing par- inder, and carefully leveling off the powder without
adigm for determination of true density as a small compacting it. The untapped apparent volume is then
amount of material is used (usually 1–8 g), the method read to the nearest graduated unit. As most phar-
is easy, reproducible and reliable, and the method is maceutical powders have densities in the range of
non-destructive: that is the material may be reused 0.1–0.7 g/mL, a 25-mL graduated cylinder filled at
after testing is complete. least 60% full calls for a sample mass of approximately
The true density, ρ, can be calculated using 2–11 g. (Since this test is non-destructive, the material
Equation 8.1. may be reused.) USP requirements dictate a minimum
w graduated cylinder size of 25 mL.17 However, if mate-
ρ (8.1) rial is in short supply, a 10-mL graduated cylinder
Vp
may be used. Although wall effects could be observed,
where: this approach provides a reasonable estimate of the
w is the weight of the sample bulk density.
Vp is the powder volume. Bulk density is an essential parameter for process
development and solid dosage manufacturing. It is
Since the measured density is a volume-weighted used in determining the amount of powder that can
average of the densities of all the individual powder fit in a space, such as a blender or a hopper on a tablet
particles, errors may be introduced if the gas sorbs press or capsule filler. It is also used to determine the
onto the powder or if volatile contaminants escape amount of powder that can be fitted into a capsule.
from the powder during the measurement. Sorption Previous work has suggested that the effective bulk
may be prevented by using an appropriate gas, usu- density of the same material will vary under different
ally helium or nitrogen. Volatile components may be dynamics.14,15
removed during purging of the sample, and sample
weight is taken before and after purging to determine
if volatile contaminants were removed. True density
is an essential parameter for process development Tapped Density
and solid dosage manufacturing. As discussed in Tapped density of a powder is the ratio of the mass
more detail later in the chapter, true density is used to of the powder to the volume occupied by the powder
calculate solid fraction. after it has been tapped for a defined period of time.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.3 POWDER CHARACTERIZATION 169

The tapped density of a powder represents its random The influence of physical and mechanical properties
dense packing. Tapped density can be calculated using on powder flow is a subject of interest to formulation
Equation 8.3, where M  mass in grams, and Vf  the scientists. Factors such as particle size distribution and
tapped volume in milliliters. particle shape have been shown to influence flow.19,20,21
Additional properties such as bulk and tapped density,
M bonding index, and internal friction coefficient are also
Tapped Density(g/mL )  (8.3)
Vf thought to be contributors. An understanding of the
effects of physical and mechanical properties on pow-
Tapped density values are generally higher for der flowability can decrease the need to perform pow-
more regularly shaped particles (i.e., spheres), as com- der flowability analysis on some materials, resulting in
pared to irregularly shaped particles such as needles. significant time and resource savings.
Particle size distribution has been shown to affect the An attempt has been made to model flowabil-
packing properties of fine powders.16 The packing ity based on physical and mechanical properties
properties of a powder can affect operations critical to using complex methods such as artificial neural net-
solid dosage manufacturing, including bulk storage, works (ANNs), discrete element method (DEM), and
feeding, and compaction. constitutive models.22,23,24 While these models have
Tapped density is measured by first gently intro- demonstrated a correlation between certain physical
ducing a known sample mass into a graduated cyl- properties and the results of various methods for meas-
inder and carefully leveling off the powder without uring flow, more work is required.
compacting it. The cylinder is then mechanically
tapped by raising the cylinder and allowing it to drop
under its own weight using a suitable mechanical Compressibility Index and Hausner Ratio
tapped density tester that provides a suitable fixed The compressibility index (CI) is a measure of the
drop distance and nominal drop rate. There are two propensity of a powder to consolidate.25 As such, it is a
methods for measuring tapped density in the USP measure of the relative importance of inter-particulate
with different drop distance and drop rates.17 The tap interactions. In a free-flowing powder, such interac-
density of a number of pharmaceutical materials is tions are generally less significant, and the bulk and
shown in Figure 8.4.18 As with bulk density measure- tapped densities will be closer in value. For poorer
ments, a material-sparing approach can be undertaken flowing materials, there are frequently greater inter-
by using a 10-mL graduated cylinder (1–4 gram sam- particle interactions; bridging between particles often
ple requirement). This test is also non-destructive. results in lower bulk density and a greater difference
between the bulk and tapped densities. These differ-
ences in particle interactions are reflected in the CI. A
8.3.2 Flow general scale of powder flow using the CI is given in
Table 8.3.25 Compressibility index (CI) can be calcu-
Flow assessment of active pharmaceutical ingredi-
lated as shown in Equation 8.4, where Vo  untapped
ents (APIs), excipients, and formulations are routinely
apparent volume, Vf  tapped apparent volume.
completed as part of solid dosage form develop-
ment. Assessments must be made to ensure powder (Vo  V f )
will flow adequately through processing equipment CI(%)  100
(8.4)
Vo
such as a roller compactor, hopper or tablet press.
Poor flowability can lead to the inability to feed pow- Although this method cannot be used as a sole
der into the dies of a rotary tablet press, and can also measure of powder flowability, it has the advantage of
cause tablet weight variation. being simple to calculate, and it provides a quick com-
Due to the complexity of powder flow, and the fac- parison between API, excipients, and formulations. If
tors that influence it, no single measure is currently bulk and tapped density measurements have already
adequate for defining flow. Unsurprisingly, many been performed, no additional material or experimen-
ways to measure flow currently exist, ranging from tation is required to calculate the CI. CI has been cor-
simple, qualitative methods, to more quantitative related to manufacturing performance on machines
methods utilizing specialized technology. Factors such such as capsule fillers. Podczeck et al. demonstrated
as the relative humidity of the environment, previous a correlation between the minimum coefficient of fill
storage conditions, and degree of consolidation have a weight variation and CI.26
large impact on flowability, any of which can alter the The Hausner ratio (HR) is closely related to
test results. CI.27 It can be calculated using Equation 8.5, where

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


170 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

60

50

40
Compressibility index

Powder
30 Mag stearate
Talc
Dical
20 Sucrose
B Lactose
Corn starch
MCC-Med
10
MCC-Coarse
FT lactose
NaCl
0
0 1000 2000 3000 4000 5000 6000 7000
Number of Taps
FIGURE 8.4 Influence of number of taps on the compressibility index

TABLE 8.3 Scale of flowability for compressibility index and Due to the high dependence of angle of repose meas-
Hausner ratio31
urements on testing conditions, angle of repose is not
Compressibility a very robust means of quantifying powder flow.31
Flow character index Hausner ratio Flow rate through an orifice is generally measured as
the mass of material per unit time flowing from any of a
Excellent 10 1.00–1.11 number of types of containers (cylinders, funnels, hop-
Good 11–15 1.12–1.18 pers). It is thought to be a more direct measure of flow
Fair 16–20 1.19–1.25 than measurements such as angle of repose or Hausner
Passable 21–25 1.26–1.34 ratio, because it more closely simulates flow of material
Poor 26–31 1.35–1.45 from processing equipment such as from a tablet press
Very poor 32–37 1.46–1.59 hopper into a die. Measurement of the flow rate is heav-
Very, very poor 38 1.60
ily dependent on test set-up, such as orifice diameter.32
Both angle of repose and flow-rate through an orifice
methods require 5–70 grams of material, and therefore
are not aligned with material-sparing strategies.
Vo  untapped apparent volume and Vf  tapped
apparent volume.
Shear Cell Methods
Vo
Hausner Ratio  (8.5) Shear cell methods measure flow on a more fun-
Vf
damental basis than the simple methods discussed
Scales of flowability for compressibility index and above, providing more robust flow results. Shear cell
Hausner ratio are included in Table 8.3. methods allow the assessment of flow properties as
a function of consolidation load and time, as well as
powder–hopper material interactions.33 Various types
Angle of Repose and Flow Through an Orifice of shear cells exist, including rotational and transla-
The angle of repose has long been used to charac- tional cells.34,35,36 While shear cell measurements are
terize bulk solids.28,29,30 Angle of repose is a character- generally more time-consuming than the methods
istic related to inter-particulate friction or resistance to discussed above, they offer a higher degree of experi-
movement between particles. According to the USP, mental control, leading to more reproducible results.
it is the constant, three-dimensional angle (relative to They are used extensively in multiple industries, and
the horizontal base) assumed by a cone-like pile of significant advances in automation have occurred in
material formed by any of several different methods. the past five years.37,38,39

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.3 POWDER CHARACTERIZATION 171

Yield loci for three materials TABLE 8.4 Physical and mechanical properties and
3000
environmental factors influencing flowability
n s
Sucrose 1.56 45. Physical and mechanical
properties Environmental effects
Bolted lactose 1.49 54.
Spray dried lactose 1.02 22.
Particle size distribution Consolidation load
2000 Particle shape Consolidation time
τ, kdynes

Bulk/tapped density Shear effects (direction & rate)


Particle density Particle/wall interactions
Moisture content Air permeability
Crystallinity Compressibility (a function of
1000 load and time)
Polymorphic form Humidity/powder moisture
content
Surface area Packed density
Electrostatic charge
Surface energy
0 1000 2000 3000 Elasticity
σ, kdynes Plasticity (ductility)
FIGURE 8.5 Yield loci for three materials Viscoelasticity
Brittleness

The shear cell analysis method is as follows: a bulk


solid sample is carefully loaded into a container or representations of the relationship between the nor-
“cell,” taking care not to consolidate the powder during mal and shear stresses. Mohr stress circles represent
loading. The powder bed is typically preconsolidated at the stresses in cutting planes of a bulk solid, which
a defined load normal to the powder bed. As the load are inclined through all possible angles. These stress
is applied, the powder bed is sheared until a uniform circles are defined by Equation 8.6 and Equation 8.7,
state of consolidation is reached (i.e., powder starts to where σy is the stress acting on the element of bulk
“flow” and shear stress reaches a constant value). This solid in the vertical direction, σx is the stress prevail-
value of the shear stress represents the shear strength ing in the horizontal direction as a result of the vertical
of the powder at those conditions. Once preconsolida- stress, and α is the angle defined by the cutting plane.
tion is achieved, the normal load is reduced, and a nor-
mal load less than the preconsolidation load is applied. (σ y  σ x ) (σ y  σ x )
The bulk solid is sheared until the shear force goes σn   cos(2α ) (8.6)
2 2
through a maximum value and then begins to decrease.
This maximum value represents the shear strength of
⎛ σ y  σ x ⎞⎟
the powder bed under those conditions. This process τ  ⎜⎜⎜ ⎟⎟ sin(2α ) (8.7)
of preconsolidation, and subsequent consolidation at ⎜⎝ 2 ⎟⎠
a reduced load, is repeated multiple times at different
normal loads. This process is sometimes referred to as A “small” Mohr stress circle is constructed tan-
a yield locus test. A yield locus is constructed based on gential to the yield locus with its minor principle
the shear data; it passes through the points defined by stress equal to zero. The “larger” Mohr stress circle
the shear and normal stress values during pre-shear is constructed tangential to the yield locus, and run-
and shear. Figure 8.5 shows the yield loci of three dif- ning through the pre-shear point. The larger Mohr
ferent materials, constructed using a simplified (trans- stress circle represents the stress state in the bulk
lational) shear cell. The yield locus is characteristic of solid sample at steady state flow (i.e. the stress state
the physical and mechanical properties of the powder, at the end of pre-shear). These two Mohr stress cir-
as well as a number of environmental factors (a list of cles help define the flow properties listed in Table
some of these factors is included in Table 8.4). 8.5.40 An example of a yield locus and Mohr stress
In addition to the yield locus, Mohr stress circles circles is shown in Figure 8.6. Some of the most com-
are constructed from the shear data to create graphical monly reported shear cell flow properties reported in

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


172 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

TABLE 8.5 Shear cell flow property names and definitions the literature are the angle of internal friction and the
flow function.41,42
Flow property Definition
Because shear methods provide quantitative, repro-
ducible results, the properties defined in Table 8.5 can
Major consolidation Defined by the major principal
stress (σ1) stress of the larger Mohr stress be used to compare the flowability of different drug
circle. substances, excipients, and formulations. To prop-
Unconfined yield Defined by the major principle erly compare data, the environmental factors listed
strength/stress (σc) stress of the smaller Mohr stress in Table 8.4 should be kept constant. For example,
circle. It is the stress at which the Moreno-Atanasio et al. demonstrate an increase in
sample will break (flow) after a the unconfined yield strength with an increase in
vertical stress has been applied to
a consolidated sample.
preconsolidation stress using a uniaxial compression
test. Shear cell methods are also used to character-
Flow function coefficient The ratio of the major
(FFC) consolidation stress to the ize the performance of materials on equipment such
unconfined yield strength at a as capsule-filling machines,43 and tablet presses.44
defined normal load. They are extensively used to design hoppers for feed-
Flow function A flow function curve can be ing powders, and to study the effect of storage time
constructed by plotting the FFC on flowability.45,46 Jenike developed a mathematical
values obtained by performing methodology for determining the minimum hopper
multiple yield locus tests at
different preconsolidation normal
angle and opening size for mass flow from conical
loads. and wedge shaped hoppers.47
Slope angle of the linearized The angle defined by the Several different types of shear cell testers are avail-
yield locus (ϕlin) linearized yield locus. The able. The Schulze Ring Shear Tester is an annular or rota-
linearized yield locus is the line tional shear tester. The bulk solid sample is contained
that is tangential to both Mohr in an annular shear cell containing a ring at the bottom
stress circles.
and a baffled lid. Ring shear testers are considered to be
Effective angle of internal The angle defined by the line that relatively easy to operate, and provide good reproduc-
friction (ϕe) runs through the origin of the
diagram, and is tangent to the
ibility.40 A single yield locus test can be performed in
larger Mohr stress circle. approximately 30 minutes (of which the operator must be
Angle of internal friction at The arctangent of the ratio of present about one third of the time). Multiple shear cell
steady state flow (ϕsf) shear stress to normal stress of sizes are available. Depending on the particle size of the
the preshear point (i.e., steady powder specimen a small, 10-mL shear cell can be used,
state flow). It characterizes the allowing for a significant reduction in sample require-
internal friction at steady state
ments. Medium and large shear cells are also available,
flow in the shear plane.
which are able to accommodate samples with larger par-
Cohesion (τc) The shear stress at yield under
zero normal stress, i.e. the
ticle sizes. These cell sizes require sample volumes greater
intersection of the yield locus than approximately 31 mL and 73 mL, respectively.48 A
with the ordinate. standard test method for using the Schulze Ring Shear
Tester can be found in ASTM Method D 6773.49
The Jenike Shear Tester is a translational tester. The
shear cell is cylindrical, and is split horizontally, forming
a shear plane between the lower stationary base and the
upper moveable portion. It consists of a closed ring at the
bottom, a ring of the same diameter lying above the bot-
Effective yield locus
tom ring, and a lid.52 An advantage of the Jenike cell over
the ring shear tester is that the powder bed is sheared
more uniformly.52 Disadvantages of the Jenike Shear
Linearized yield locus Tester include the amount of material required, and the
tau

time required for a test. Depending on the powder, and


the operator’s skill, one to two hours per yield locus (dur-
ϕlin ing which the operator has to be present) are required.
ϕe Results generated using the Jenike Shear Tester have been
τc shown to correspond to those generated using the ring
σe Sigma σ.1 shear tester.40 A standard test method for using the Jenike
FIGURE 8.6 Yield locus and Mohr stress circle diagram Shear Tester can be found in the ASTM Method D 6128.50

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.4 COMPACT (MECHANICAL PROPERTY) CHARACTERIZATION 173

Additional Shear Testers Bhattachar et al. developed a vibratory feeder


Schulze ring shear and Jenike shear cell testers are method for assessing avalanche behavior that requires
used extensively due to their commercial availability, a smaller sample size than for the Aeroflow® (1.2 g).
though other shear cells are available. Plate or “simpli- Results compared to those generated using the
fied” shear cells have also been designed, which consist Aeroflow®.60 Despite the small sample size require-
of a thin sandwich of powder between a lower station- ments this method is not widely used. The instrument
ary rough surface and an upper rough surface that is is not commercially available, and has not been exten-
movable.18,51 Uniaxial, biaxial, and triaxial testers have sively tested.
also been used for flow analysis, and have been dis-
cussed in the literature. The measurement principle of
the uniaxial tester is similar to that of shear cells.35,52 8.4 COMPACT (MECHANICAL
PROPERTY) CHARACTERIZATION
Dynamic Test Methods
Avalanche testers assess the flowability of powders Many investigations have demonstrated the impor-
by measuring their avalanching behavior, which is tance and impact of the physical and chemical prop-
related to powder cohesivity and flowability. Unlike erties of materials on powder processing. Physical
shear cell methodology, this type of assessment is properties such as particle size and shape clearly
dynamic in nature,53 which may be more applicable influence powder flow for example. The previous
to low-shear processes such as blending, in which sections of this chapter provide some recommenda-
avalanching behavior of powder promotes mixing. tions for how to proceed with characterization using
Avalanche testing can be carried out in different types limited quantities of materials. However, compact
of equipment, including rotating drums and vibratory mechanical properties (i.e., those properties of a mate-
feeders.54,55 Avalanche testing has been shown to dis- rial under the influence of an applied stress) are also
tinguish between freely flowing powders, blends, and of great importance for solid dosage form develop-
granulations.56,57 ment and manufacturing—particularly for tablet for-
One of the most widely used rotating drum ava- mulation. This section describes the importance of the
lanche testers is the Aeroflow® (TSI, St. Paul, MN). mechanical properties of materials, as well as some
Powder is filled into a transparent drum, which is basic principles and methodologies that can be used
then rotated at a fixed speed. The stress applied to to investigate the influence of these properties on
the powder sample as a result of rotation causes the compaction. For the purposes of this discussion, phys-
powder to shear, resulting in avalanche events.58 The ical properties are considered to be those properties
avalanche events are monitored by an optical sensor that are “perceptible especially through the senses”
system. From the detector response data, a frequency (i.e., properties such as particle size, and shape). In
or mass distribution of the avalanche events can be contrast, mechanical properties are those properties of
generated which can be used to determine various a material under an applied load: elasticity, plasticity,
flowability parameters. Hancock et al. used the mean viscoelasticity, bonding, and brittleness.
time to avalanche (mean of the distribution), and the Table 8.4 lists some of the physical and mechani-
coefficient of variation of the avalanche events, to cal properties that influence powder properties and
characterize powder flow.54 compaction. For example, surface energy and elastic
One limitation of the avalanche tester is the quali- deformation properties influence individual parti-
tative nature of determining the regimes of avalanche cle true areas of contact. Plastic deformation likely
flow in the rotating drum. Boothroyd et al. argued that occurs to some extent in powder beds depending on
the ideal flow regimes necessary for meaningful data the applied load, and almost certainly it occurs dur-
analysis for pharmaceutical powders were the “rolling” ing the compaction of powders into tablets. Certainly
and “cascading” regimes.59 Another limitation of the at asperities, local regions of high pressure can lead to
system is the method development required prior to localized plastic yielding. Electrostatic forces can also
analysis. Rotational speed, measurement duration, and play a role in powder flow, depending on the insulat-
sample size must be optimized, as they have a great ing characteristics of the material and environmental
degree of influence over the measurement results.54 conditions. Particle size, shape, and size distribution
A third limitation is the amount of material required, have also been shown to influence flow and com-
which limits the use of this technique in a material- paction. A number of environmental factors such as
sparing approach to formulation development. Hancock humidity, adsorbed impurities (air, water, etc.), con-
et al. proposed a sample size of 50 mL.54 solidation load and time, direction and rate of shear,

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


174 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

and storage container properties are also important. Elastic Deformation


With so many variables, it is not surprising that a In general, during the initial stages of deformation,
wide variety of methods have been developed to char- a material is deformed elastically. A change in shape
acterize materials. The focus of this chapter is on those caused by the applied stress is completely reversible,
useful methods that require limited amounts of mate- and the specimen will return to its original shape on
rial (bulk drug or formulation), and provide the most release of the applied stress. During elastic deforma-
valuable information. tion, the stress–strain relationship for a specimen is
What holds particles together in a tablet? A detailed described by Hooke’s law (Equation 8.8):
discussion is beyond the scope of this chapter, and
excellent references are available in the literature.61,62 σ  E
ε (8.8)
However, it is important to realize that the forces that
hold particles together in a tablet or powder bed are where:
the very same forces discussed in detail in introduc- E is referred to as Young’s modulus of elasticity
tory physical chemistry texts. There is nothing magical σ is the applied stress
about particle–particle interactions; the forces involved ε is the strain (ε  (l  lo )/lo ).
are London dispersion forces, dipole interactions, sur- The region of elastic deformation of a specimen is
face energy considerations, and hydrogen bonding. The shown graphically in Figure 8.6. The reader is directed
consolidation of powders brings particles into close to standard texts in material science and engineer-
proximity, and these fundamental forces can begin to ing for detailed discussions of elastic deformation. As
act effectively to produce strong particle–particle inter- long as the elastic limit is not exceeded only elastic
actions (e.g., bonding). Particle rearrangement, elastic deformation occurs.
and plastic deformation of material can establish large The elastic properties of materials can be under-
areas of true contact between particles; if the resulting stood by considering the attractive and repulsive
particle–particle bonds are strong, a strong and intact forces between atoms and molecules. Elastic strain
tablet is produced. results from a change in the intermolecular spacing
and, at least for small deformations, is reversible.

8.4.1 Important Mechanical Properties


Plastic Deformation
Materials used in the pharmaceutical industry can Plastic deformation is the permanent change in
be elastic, plastic, viscoelastic, hard, tough or brittle shape of a specimen due to applied stress. The onset
in the same sense that metals, plastics or wood are. of plastic deformation is seen as curvature in the
The same concepts that mechanical engineers use to stress–strain curve shown in Figure 8.7. Plastic defor-
explain or characterize tensile, compressive or shear mation is important because it “allows” pharmaceuti-
strength are relevant to pharmaceutical materials. cal excipients and drugs to establish large true areas of
These mechanical properties of materials can have a contact during compaction that can remain on decom-
profound effect on solids processing. pression. In this way, strong tablets can be prepared.
The mechanical properties of a material play an
important role in powder flow and compaction. These
properties are critical properties that influence the Elastic
true areas of contact between particles. Therefore, it deformation
Plastic
is essential to characterize the properties. Reliable Ultimate tensile
mechanical property information can be useful in strength
helping to choose a processing method such as granu-
lation or direct compression, selecting excipients with X fracture
Stress, σ

properties that will mask the poor properties of the


drug or helping to document what went wrong, for
Elastic limit
example, when a tableting process is being scaled-
up or when a new bulk drug process is being tested.
Since all of these can influence the quality of the final
product, it is to the formulator’s advantage to under-
stand the importance of the mechanical properties of
the active and inactive ingredients, and to be able to Strain, ε
quantify the properties. FIGURE 8.7 Stress–strain curve

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.4 COMPACT (MECHANICAL PROPERTY) CHARACTERIZATION 175

Plastic deformation, unlike elastic deformation, speed is rapid. All pharmaceutical materials are vis-
is generally not accurately predicted from atomic or coelastic; the degree to which their mechanical prop-
molecular properties. Rather, plastic deformation erties are influenced by rate depends on the material.
is often determined by the presence of crystal defects
such as dislocations, grain boundaries, and slip
planes within crystals. While it is not the purpose of
8.4.2 Overview of Methods
this chapter to discuss this in detail, it is important
to realize that dislocations and grain boundaries are Characterizing mechanical properties has been an
influenced by factors such as the rate of crystalliza- active area of pharmaceutical research for decades. The
tion, particle size, the presence of impurities, and application of classic “engineering” methodologies
the type of crystallization solvent used. Slip planes to characterize pharmaceutical materials dates to the
may exist within crystals due to molecular packing 1950s or before. With the advent of high-speed compu-
arrangements that result in weak interplanar forces. ter control, and monitoring of processes such as tablet
Processes that influence these (e.g., crystallization rate, compaction, the era of “dynamic” characterization of
solvent, temperature) can be expected to influence the pharmaceutical materials was ushered in. Sophisticated
plastic deformation properties of materials, and hence instrumentation of rotary tablet presses and, in par-
the processing properties. The reader is directed to ticular, the design of tablet compaction simulators with
standard texts in material science and engineering for seemingly infinite control of the compaction process,
detailed discussions of plastic deformation. has offered scientists an unprecedented opportunity to
The plastic properties of a material are often study the mechanics of materials at speeds representa-
determined by an indentation test.63 Both static and tive of production tablet compaction. Yet, even today,
dynamic test methods are available, but all generally both dynamic testing and the classic “quasi-static”
determine the pressure necessary to cause permanent engineering testing approaches offer opportunities to
and non-recoverable deformation. understand pharmaceutical materials. In this regard,
dynamic and quasi-static testing are complementary
tools. Both quasi-static and dynamic test methodolo-
Brittle and Ductile Fracture gies will be discussed in the following sections. One
In addition to plastic deformation, materials may key advantage of quasi-static testing is the ability to
fail by either brittle fracture or ductile fracture; frac- “independently” dissect out and investigate the vari-
ture being the separation of a body into two or more ous mechanical properties of a material. As stated pre-
parts. Brittle fracture occurs by the rapid propaga- viously, pharmaceutical materials can be elastic, plastic,
tion of a crack throughout the specimen. Conversely, viscoelastic, hard, tough or brittle. Ultimately, these
ductile fracture is characterized by extensive plastic individual components that cumulatively describe
deformation followed by fracture. Ductile failure is a pharmaceutical material determine its compaction
not typically seen with compacts of pharmaceutical properties in a dynamic compaction process.
materials. The characteristic snap of a tablet during The consolidation of powders into intact tablets is a
hardness testing is indicative of brittle fracture. process of reducing pores in a powder bed while creat-
ing interparticle bonds. During compression, materials
experience complex stresses, the structure of the pow-
Viscoelastic Properties der bed changes, and consolidation is brought about
Viscoelastic properties can be important; viscoe- mainly by particle rearrangement, plastic deformation,
lasticity reflects the time-dependent nature of stress– and fragmentation.64 The deformation of pharmaceuti-
strain. A basic understanding of viscoelasticity can be cal materials is time dependent, and this dependency
gained by considering processes that occur at a molec- is related to the consolidation mechanism and dynam-
ular level when a material is under stress. An applied ics of the consolidation process.65,66,67,68,69 Under com-
stress, even when in the elastic region, effectively pression, for example, brittle materials are considered
moves atoms or molecules from their equilibrium to consolidate predominantly by fragmentation; plas-
energy state. With time, the rearrangement of atoms or tic materials deform by plastic flow. The time depend-
molecules can occur. ency of this process arises from stress relaxation for
The stress–strain relationship can therefore depend materials undergoing primarily plastic deformation.
on the time frame over which the test is conducted. In However, the compaction of brittle materials is often
compacting tablets, for example, it is frequently noted less influenced by speed, because fragmentation is rap-
that higher compaction forces are required to make idly achieved and prolonged exposure to the force has
a tablet with a given strength when the compaction a limited effect on tablet properties.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


176 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

Several researchers have previously identified the may exist in the test specimen itself. Of the methods
utility of solid fraction in describing tablet properties. defined in the literature, the most refined method is
Armstrong and Palfrey70 concluded that differences in to make square compacts using triaxial compression
the tensile strength of tablets compressed at different and decompression.74 A split die (Figure 8.8) is used
speeds could be accounted for by differences in tablet to make compacts that are substantially free of defects
porosity. Hancock and coworkers71 found that tablet that may occur if a conventional compaction process
strength and disintegration time for tablets made on an were to be used. The split die permits triaxial decom-
eccentric press and a rotary press were comparable when pression such that the pressure applied to all three
considering a comparable solid fraction. Maarschalk and axes is essentially equal during the decompression
coworkers72 found that tablet tensile strength of sorbitol process.74 This is achieved by computer control of the
as a function of tablet porosity was independent of com- decompression process. The stresses in the compact
pression speed. Finally, Tye and coworkers64 extended are more uniformly relieved in three dimensions, and
this work to show that tablet solid fraction (SF) was the this minimizes the production and propagation of
primary factor determining tablet strength for several flaws within the compact.
pharmaceutical excipients (both brittle and ductile) over
an extremely wide range of compaction speeds (dwell
Importance of the Solid Fraction
times from 10 msec to 90 sec).
The solid fraction (SF) of a compact can be calcu- It is imperative to realize and address the fact that
lated based on the true density (ρtrue) of the material the mechanical properties of a compact are very much
(typically determined using pycnometry), the tablet influenced by solid fraction. A change in solid frac-
volume (ν), and the tablet weight (Wt) (Equation 8.9): tion of 0.01 (i.e., a change in SF from 0.85 to 0.86) can
result in a mechanical property change of 10% to 20%.
Wt
SF  (8.9) For this reason, it is critical to compare the properties
ρtrue
ν of a material at a “reference” solid fraction to ensure
that one is “comparing apples to apples.” Hiestand
The relationship between the solid fraction, also
and coworkers74 defined their reference solid fraction
referred to as relative density, and porosity (ε) is:
as 0.9 (i.e., porosity  0.1 or 10%) while others have
ε  1  SF (8.10) used a solid fraction of 0.85 or even extrapolated to
a solid fraction of 1.0 (e.g., zero porosity). In compar-
ing results from the literature, it is important to keep
this in mind. It is recommended that a solid fraction
8.4.3 Quasi-static Testing in the range typical of tablet compaction be used.
Quasi-static testing typically applies variations of tra- For compacts of organic materials, a reference solid
ditional engineering and material science testing meth- fraction of 0.85 is in the midrange of those typically
ods to compacts (i.e., test specimens) of pharmaceutical
materials. There are, for example, a number of variations
of indentation, tensile, flexural, compression, and brit-
tle fracture tests in the pharmaceutical literature.73 The Side view I
quantity of material required for testing varies from 1 to
100 grams. Methods for characterizing the elastic, plas- Top view
D
B D
tic, and brittle properties of compacts of organic materi-
als have, for example, been developed by Hiestand and C A C
coworkers.74,75,76,77,78 These measures of tableting per-
F
formance assess several key mechanical properties of
compacted materials that have been shown to relate to
H G
tableting. Currently available methods typically require
from 10 to 60 grams for complete mechanical property
characterization using Hiestand’s methods. E

F J

Test Specimen Preparation


It is important to properly prepare test speci-
mens of pharmaceutical materials so quasi-static test FIGURE 8.8 Schematic drawing of a simple triaxial press with
results are not improperly influenced by “flaws” that a split die

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.4 COMPACT (MECHANICAL PROPERTY) CHARACTERIZATION 177

observed. For inorganic materials (such as dicalcium pharmaceutical excipient. One can clearly see the
phosphate) solid fractions in the 0.6 to 0.75 range are impact of solid fraction on the measured mechanical
often observed for tablets. The wide range of mechan- properties.
ical properties observed means no ideal value can be
identified for all materials.
Tableting Indices
Using the methodology described above, several
Tensile Strength Determination indices of tableting performance have been developed
The tensile strength, σT, of a square test specimen pro- by Hiestand and coworkers.61,76 These indices provide
vides extremely useful information. Several methods relative measures of properties (i.e, dimensionless
for determining it are available, and include traditional numbers) that reflect the performance of materials
tablet hardness testing and transverse compression or during processing.
square compacts. In transverse compression, specimens
are compressed with platens 0.4 times the width of the
compacts in the tensile testing apparatus.74 The force
necessary to cause tensile failure (tensile forces are maxi-
mum at the center of the tablet) is monitored by a load
cell, and the magnitude of the force at fracture is deter-
mined. Testing of square compacts has advantages over
the testing of circular compacts; however, circular com-
pacts can be used. Conventional hardness testing of
tablets can result in a measurement of tensile strength.79
Similar results are obtained for round and square com-
pacts when tensile failure is achieved. It is extremely
important to compare measured properties such as ten-
sile strength at the same solid fraction. Tensile strength
values in excess of 1 MPa (typical range 0.1 to 4 MPa) are
typically desired for tablets.

Pendulum Impact Device


A simple schematic of a pendulum impact device
(PID) is given in Figure 8.9. This equipment per-
mits the permanent deformation pressure of a com-
pact of material to be determined under dynamic
conditions.74,75 Flat-faced, square tablets of the test FIGURE 8.9 Pendulum impact device
substance are compressed at different compression
pressures, and then subjected to impact with a stain-
less steel ball in the PID. The rebound height of the
ball and the chordal radius of the dent are carefully Reference solid fraction
Dynamic hardness (MPa) or tensile

measured, and used to calculate the permanent defor- 1000


mation pressure. In a simple sense, one is measuring
the energy necessary to make the permanent defor-
strength (MPa)

mation (the difference between the initial height of 100


the ball and the rebound height). By measuring the Dynamic hardness
volume of the dent, one can calculate the deforma- Tensile strength
tion pressure—the energy divided by the volume. The 10
permanent deformation pressure is the pressure (i.e.,
stress) necessary to cause plastic deformation. This
permanent deformation pressure, H, has been shown 1
to be related to the yield pressure obtained using 0.7 0.75 0.8 0.85 0.9 0.95
dynamic testing methods and Heckel analysis.80 The Solid fraction
dynamic hardness and tensile strength are shown in FIGURE 8.10 Dynamic hardness and tensile strength as a func-
Figure 8.10 as a function of solid fraction for a common tion of solid fraction

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


178 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

Bonding index ⎡σ ⎤
BFI  0.5
⎢ T  1⎥ (8.12)
The purpose of the bonding index is to estimate ⎢σ ⎥
⎣ To ⎦
the survival of strength during decompression;76 it is
defined in Equation 8.11:
Viscoelastic index
σT
BI  (8.11) Hiestand and coworkers have further refined the
H
concept of bonding index to include both a worst-case
where: and a best-case bonding index.76 The bonding index
σT is the tensile strength of the compact at a given is determined under different experimental condi-
solid fraction (typically 0.85 or 0.9 as defined by the user) tions: the rate at which the permanent dent is made in
H is the permanent deformation pressure (i.e., hard- a compact is varied such that the viscoelastic proper-
ness) of a compact at the same solid fraction. ties of the material are assessed. If a material is very
viscoelastic, there is substantial stress relaxation with
The bonding index (BI) is, in essence, a measure of
time. It is reasonable to expect, then, that tablets that
the ability of a material, on decompression, to main-
are slowly deformed during the determination of
tain a high fraction of the bond that was created dur-
the hardness, H, may retain more of the bonded area
ing compression. At maximum compression pressure,
than tablets that are rapidly deformed (i.e., as in the
the bonded areas are at a maximum, because the
pendulum impact device), since some of the stresses
true areas of contact are maximized. During decom-
developed during compaction will have a chance to
pression, some of that area and bond is “lost” due to
be relieved. The dynamic bonding index (BId), some-
elastic recovery. A high bonding index indicates that,
times called the worst case BI, is determined using a
relatively speaking, a larger portion of the strength
the pendulum impact device (PID) for measuring the
remained intact after decompression. A low bond-
indentation hardness (Hd), while the quasi-static bond-
ing index indicates that less of the strength remains.
ing index (BIqs), also sometimes referred to as the best
The term bonding index, then, is a good description
case BI, is measured using a “quasi-static” or slow
since it, in effect, characterizes the tendency of the
method for measuring indentation hardness (Hqs). The
material to remain intact after it has been compressed.
dynamic and quasi-static bonding index is calculated
Tablets made of materials with poor bonding char-
as previously described. The viscoelastic index (VE)
acteristics may be quite friable. Compacts made of
is defined as the ratio of the dynamic to quasi-static
materials with good bonding indices may, conversely,
indentation hardness:
make strong tablets. A bonding index in excess of 0.01
(range 0.001 to 0.06) is typically desired. Hd BI qs
VE   (8.13)
H qs BI d
Brittle fracture index
The brittle fracture index is a measure of the brit-
tleness of a material. It is a measure of the ability of Application of Quasi-static Testing to
a compact to relieve stress around compact defects by Formulation Development
plastic deformation. The brittle fracture index (BFI)
The application of quasi-static testing methods and
is determined74,75 by comparing the tensile strength
interpretation has been discussed extensively in the
of a compact, σT, with that of a compact with a small
scientific literature. In addition to the pioneering work
hole (stress concentrator) in it, σTo, using the tensile
of Hiestand and coworkers,61,62,63,74,75,76,77,78 additional
test described above. A hole in the center of a com-
research discussing the application of this methodol-
pact weakens it. If a material is very brittle, theoreti-
ogy is available.33,64,81,82,83,84,85,86,87,88,89,90 Benefits of a
cal considerations show that the tensile strength of
complete characterization of the mechanical proper-
a tablet with a hole in it will be about one-third that
ties of both the active ingredient and the excipients
of a “defect free” tablet. If, however, the material can
used in the formulation include:
relieve stress, then the strength of the compact with a
hole in it will approach that of a compact with no hole. ● fundamental understanding of critical mechanical
The brittle fracture index is defined such that very properties of the active ingredient and excipients;
brittle compacts have a BFI of 1, and very nonbrit- ● identification of mechanical property deficiencies
tle materials have a BFI close to 0; it is calculated in and attributes;
Equation 8.12.74 BFI values less than 0.3 (range 0 to 1) ● selection of excipients that can overcome
are indicative of relatively nonbrittle materials. deficiencies of active ingredient;

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.4 COMPACT (MECHANICAL PROPERTY) CHARACTERIZATION 179

● identification of lot-to-lot variations in materials; While a simplification, this approach has been used
● identification of potential manufacturing problems to predict the properties of mixtures.91,92,96 An exam-
associated with tableting process. ple of the predicted mechanical properties of a ternary
The reader is directed to the literature for a thorough blend of API, and a placebo component consisting of
discussion of the application of mechanical property microcrystalline cellulose and lactose spray process, is
characterization to formulation development. A fun- shown in Figure 8.12. A sound understanding of the
damental understanding of the mechanical properties mechanical properties of the individual components,
is essential to understanding compaction properties and the range of desirable mechanical property values,
and the tableting process.61,62,76,77,78,80 Development allows for a rational selection of excipient types, grades
of mathematical models of mixtures has been used and quantities.
by Amidon,91,92 and others,88,87,93 to identify the type
and quantity of excipient required to produce tab-
let formulations that have acceptable manufacturing 8.4.4 Dynamic Testing
properties. Figure 8.11 shows mechanical properties
of binary mixtures of microcrystalline cellulose and The most commonly used methods of studying the
lactose spray process.91,92,96 From this figure, one can mechanical properties of solids under dynamic condi-
see that the mechanical properties of a mixture may tions include the use of the following instrumented
be estimated knowing the mechanical properties of equipment:
the two individual components. While the mechanical ● Hydraulic press;
properties of mixtures are complicated,94,95 estimating ● Eccentric (single station) tablet press;
the properties of mixtures has been successfully used ● Rotary tablet press;
to identify suitable excipient types and quantities. A ● Compaction simulator;
simplified equation for binary mixtures is given in ● Compaction emulator (e.g., Presster™).
Equation 8.14 or Equation 8.15:
Accurate measurements of force and distance require
Φmixture  x(ΦA )  (1  x )(ΦB ) (8.14) careful construction of equipment and placement of
instrumentation; several publications are available
log(Φmixture )  x log(ΦA )  (1  x ) log (ΦB ) (8.15) discussing these aspects.97,98 Appropriate care is also
needed during calibration, and for correction of meas-
where:
urements due to machine dimensional changes; tablet
Φmixture is the mechanical property of interest for the
punches, for example, deform elastically and these
mixture at the reference solid fraction
changes should be considered.
ΦA, ΦB are the properties of the pure components of
While any of these instrumented presses may be
the binary mixture at the same reference solid fraction
used, the most commonly used for dynamic testing
x is the fraction of component A.
is the compaction simulator. The compaction simula-
While either equation may be used, experience tor,99 because of its sophisticated control and moni-
has shown that the log-linear relationship shown in toring of the compaction process, generally offers the
Equation 8.15 often produces better predictions for greatest flexibility in compression conditions. It is
mixtures of common lactose-microcrystalline cellulose- possible, for example, to carry out compression under
based formulations. Further work is needed, however, constant velocity conditions for the compression and
to explore the science and predictability of mixtures. decompression phases (i.e., saw-tooth compression
For ternary mixtures, that is, for mixtures contain- profile)—something not possible with other presses.
ing an active pharmaceutical ingredient as one com- Also of particular note is the demonstrated utility of a
ponent and a second “placebo” component containing “linear” tablet press emulator (Presster™), that offers
two excipients, it is possible to estimate the mechani- many of the advantages of a conventional compaction
cal properties of a mixture by extending this concept, simulator in ease of use and experimental flexibility,
as shown in Equation 8.16. as well as its ability to simulate virtually any commer-
log(Φ Formulation )  y log(ΦAPI )  (1  y ) cially available rotary tablet press.100 One limitation

([x]log(ΦA )  [1  x]logΦ (8.16) of the Presster™, though it may also be viewed as a
benefit, is that it uses compression rolls of the same
where: dimensions as those on a rotary tablet press, so com-
y is the fraction of API in the blend pression profiles are limited to those described by con-
(1  y) is the fraction of the blend that constitutes ventional tablet press geometries of the tablet punch
the placebo component. tooling as it moves under the compression rolls.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


180 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

100
Hdyn  Dynamic
% MCC vs Hw Hdyn indentation
% MCC vs Sigma T hardness
10
% MCC vs BFI
% MCC vs BIw
Plot 1 Regr
σT  Tensile
1 σT
strength

0.1 BFI
BFI  Brittle
fracture index

0.01 BIdyn  Bonding


index
BIdyn

0.001 FIGURE 8.11 Mechanical properties of mix-


0 20 40 60 80 100 tures of microcrystalline cellulose and lactose
% Microcrystalline cellulose in SD lactose spray process

100 the dosage form are ultimately judged by the formula-


tor, by quality assurance, and by the consumer.
A variety of pressure–porosity equations have been
derived over the years.101,102,103 The most commonly
referenced of them is that of Heckel.109 The Heckel
1/(1-SF)

10 equation (sometimes referred to as the Athy–Heckel


equation) was derived assuming that the change in
solid fraction (i.e., relative density) with compac-
tion pressure is proportional to the porosity of the
compact. Therefore, as porosity approaches zero, the
1 change in solid fraction with compaction pressure,
0 50 100 150 dρ/dP, approaches zero. Therefore:
Compaction pressure
dρr
FIGURE 8.12 Heckel plot. ∝ ε  (1  ρr ) (8.17)
dP
And, integrating Equation 8.17, results in the classic
Heckel equation:
With proper instrumentation, “in die” or “at pres- ⎛ 1 ⎞⎟
⎜ ⎟⎟  kP  A
sure” measurements may be made during the com- ln ⎜⎜ ⎟⎟ (8.18)
⎜⎜⎝ 1  ρ r ⎠
paction process. For example, the compaction pressure
versus tablet porosity (compressibility) may be deter-
where k is a measure of the plasticity of the mate-
mined for a single tablet. This information, obtained
rial. It is related to the yield strength, Y, of a material
under these dynamic testing conditions, can be used
by Equation 8.19.104,109
to generate an “in die” Heckel plot. Alternatively,
measurements may be made on compacts after the 1
tablet is removed from the die. These are “out of die” k (8.19)
3y
or “at zero pressure” measurements. Both in die and
out of die methods have their advantages. Among the The constants in the Heckel plot are typically deter-
advantages of out of die measurements is that they mined by regression analysis of the terminal linear por-
represent the “final product” after decompression and tion of a plot of ln(1/(1  ρr)) versus P (see Figure 8.13).
ejection. The out of die properties are those by which The yield strength, Y, of the material under the dynamic

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.4 COMPACT (MECHANICAL PROPERTY) CHARACTERIZATION 181

Tensile strength vs % API required (2–10 g). In contrast, disadvantages include


7.0 the difficulty of factoring out the individual mechani-
cal property “components” that, combined, determine
6.0 how a material behaves during compaction.
The relationships between compaction pressure,
5.0 tensile strength, and solid fraction are critical to
Tensile strength (MPa)

understanding and characterizing the compaction


process. The relationship between these three param-
4.0
eters is described as:

3.0 ● Compactibility: relationship between tensile


strength and solid fraction;
2.0
● Tabletability: relationship between tensile strength
and compression pressure;
● Compressability: relationship between compaction
1.0
pressure and solid fraction or porosity;
● Manufacturability: relationship between tablet
0.0 crushing force and compression force.
0 20 40 60 80 100
% API in blend Representative compactibility, tabletability, compress-
ability, and manufacturability profiles for a compactable
0.8 0.85 0.9 excipient are shown in Figures 8.14 through Figure 8.17.
FIGURE 8.13 Predicted mechanical properties of a ternary The compactibility, tabletability, and compressability
blend of API, micro crystalline cellulose and lactose spray process. profiles form the three faces of a three-dimensional plot
as shown in Figure 8.18.64 The Presster™ compaction
emulator was used for these studies, although other
conditions of the test is a measure of the deformability. properly instrumented presses can also be used. The
In addition to yield strength, the shape of the Heckel compaction emulator was set up to emulate a Killian
plot has been used to distinguish volume reduction RST tablet machine (250 mm compression rolls) with a
mechanisms.105 Three types or families of curves are 27 msec dwell time (corresponding to 28 800 tablets/
considered to reflect materials that undergo consoli- hour) using 10 mm diameter flat-faced round punches
dation primarily by: (a) plastic deformation, (b) frag- with no precompression force.
mentation, or (c) a variation of (a) which is plastic flow Compactibility is the ability of a powder to be
with no initial particle rearrangement. transformed into tablets with a resulting strength.107 It
Additional information regarding the compaction is represented by a plot of tensile strength versus solid
process may be obtained using dynamic testing condi- fraction. The compactibility is the most valuable of the
tions, including work of compaction, work recovered dur- three properties, since it reflects the two most impor-
ing decompression, work to overcome die wall friction, tant effects of applied pressure: tablet strength, and
etc. A detailed discussion of these opportunities is beyond solid fraction. A representative compactibility pro-
the scope of this chapter, and the reader is directed to the file of an excipient is shown in Figure 8.14. If one can
literature for further information. While very valuable as achieve an acceptable tensile strength at an acceptable
a research tool, the quantitative use of pressure–porosity solid fraction with the application of pressure, a sat-
measurements and analysis beyond the determination of isfactory tablet can be produced. Compactibility plots
yield pressure does not appear to be used routinely dur- are largely independent of the process by which com-
ing formulation development and optimization. pacts are made, since only measured tablet proper-
ties (tensile strength and solid fraction) are involved.
Compactibility plots are useful as a tool to compare
Application of Dynamic Testing to Formulation formulations made on different equipment. If the for-
Development mulations are the “same” then the “same” compact-
There are a number of reports of the use of dyna- ibility plots will be obtained.64
mic testing of active ingredients and excipients in the Tabletability is the capacity of a powder to be trans-
literature. There are two key benefits of dynamic formed into a tablet of specified strength under the
testing: (1) the properties can be determined under effect of compaction pressure.107 It is represented by
dynamic conditions representing those in a production a plot of tensile strength versus compaction pressure.
environment; and (2) small quantities are typically Tabletability describes the effectiveness of the applied

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


182 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

10.00 30
9.00
8.00 25
Tensile strength (MPa)

7.00

Tablet hardness (Kp)


6.00 20
5.00
4.00 15
3.00
2.00 10
1.00
0.00 5
0.550 0.650 0.750 0.850 0.950
Solid fraction 0
0 2 4 6 8 10
FIGURE 8.14 Compactability profile using a compaction
emulator Compression force (kN)
FIGURE 8.17 Manufacturability profile using a compaction
emulator
9.00
8.00
Tablet tensile strength (MPa)

7.00 dependent. Also, at high pressures, some materials


6.00 may have lower tensile strength due to overcompac-
5.00 tion.106 Characterization of the tabletability provides
4.00 excellent insight into the compaction process and
3.00 mechanical properties of a material.
2.00 Compressability is the ability of a material to undergo
1.00 a reduction in volume as a result of an applied pres-
0.00
sure.107,108 It is a measure of the ease with which a pow-
0 20 40 60 80 100 120 140 der bed undergoes volume reduction under compaction
Compression pressure (MPa) pressure; it is represented by a plot showing the reduc-
FIGURE 8.15 Tabletability profile using a compaction emulator tion of tablet porosity (i.e., the increase in solid frac-
tion) with increasing compaction pressure (Figure 8.16).
Compressability is often described by the Heckel equa-
1.000 tion.109 Heckel plots, for example, have been widely
0.950 used to assess the mechanism of deformation, and as
0.900
a tool to estimate yield pressure. It is also well-known
that tablet porosity is an important parameter, for exam-
0.850
ple, in tablet disintegration and dissolution, since some
Solid fraction

0.800 porosity is often necessary to facilitate liquid penetra-


0.750 tion into tablets.110,111
0.700 Manufacturability, a plot closely related to tabletability,
shows the relationship between the tablet crushing force
0.650
(related to tensile strength), and compaction force (related
0.600
to compression pressure). The manufacturability profile
0.550 (Figure 8.17) is commonly considered by formulation sci-
0 20 40 60 80 100 120 140
entists since it reflects the “measured” properties of a dos-
Compression pressure (MPa)
age form during manufacturing (tablet crushing strength
FIGURE 8.16 Compressibility profile using a compaction
and compression force). In general, however, pressure and
emulator
tensile strength are preferred parameters to consider.
In summary, characterization of the compactibility,
pressure in increasing the tensile strength of the tab- tabletability, compressability, and manufacturability
let, and demonstrates the relationship between the of a formulation provides valuable information of the
cause (the compaction pressure), and the effect (the compaction process, and the prospects for a successful
strength of the compact) (see Figure 8.15). Normally, tableting process in manufacturing. Obtaining tablets
a higher compaction pressure makes a stronger tablet. with adequate tensile strength at a reasonable solid
However, this relationship is often found to be speed fraction with acceptable compression pressure is the

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.5 CONCLUSIONS 183

Compactibility References
ty 1. Brittain, H. G. (2004). Evaluation of the Particle Size Distribution of
ili
ab

Pharmaceutical Solids. Profiles of Drug Substances, Excipients, and


et
bl

Related Methodology, Vol. 31.


Ta

2. Allen, T. (1997). Particle Size Measurement, 5th edn. Vol. 1.


14 Chapman & Hall, New York.
3. Stanforth, J. (2004). Particle-size Analysis, In: Pharmaceutics The
12 Science of Dosage Form Design, M. Aulton (ed.). pp. 152–165.
4. US Pharmacopeia 27. (2006). US Pharmacopeial convention,
ngth, MPa

10 Rockville, MD, pp. 2716–2717.


5. Newman, A. W. & Brittain, H. G. (1995). Particle Morphology:
8
Optical and Electron Microscopies. In: Physical Characterization
Tensile stre

MPa
6 250
of Pharmaceutical Solids, H.G. Brittain, (ed.). Marcel Dekker,
New York. pp. 127–156.

ure,
4 200
6. Operation and Maintenance Manual. (2004). Gilsonic Siever

ress
150 Model GA-8, Gilson Company, Inc., P.O. Box 200, Lewis Center,
2

ion p
100 Ohio 43035-0200.
0 7. US Pharmacopeia 27. (2006). US Pharmacopeial convention,
pact
0.5
0.6 Compress 50
0.7 ibility Rockville, MD. pp. 2720–2722.
0.8
Com
0.9 8. Rohrs, B.R., Amidon, G.E., Meury, R.H., Secreast, P.J., King, H.M. &
Solid fract 1.0
ion
Skoug, C.J. (2006). Particle size limits to meet usp content uni-
FIGURE 8.18 Three-dimensional tablet tensile strength, solid formity criteria for tablets and capsules. Journal of Pharmaceutical
fraction, and compaction pressure curve Sciences 95(5), 1049–1059.
9. Dressman, J.B. & Fleisher, D. (1986). Mixing tank model for pre-
dicting dissolution rate control of oral absorption. Journal of
key to success. Robust formulations must not be on Pharmaceutical Sciences 75, 109–116.
the “edge;” that is, they should provide the manufac- 10. Higuchi, W.I. & Hiestand, E.N. (1963). Dissolution rates of finely
divided drug powders I: Effect of a distribution of particle sizes
turing scientist with the ability to adjust compression
in a diffusion-controlled process. Journal of Pharmaceutical
pressure to achieve the desired tensile strength and Sciences 52, 167–171.
still maintain the solid fraction in a desirable range, 11. Hintz, R.J. & Johnson, K.C. (1988). The effect of particle size dis-
such that the tablet performs as required. tribution on dissolution rate and oral absorption. International
Journal of Pharmaceutics 51, 9–17.
12. Rohrs, B. R. & Amidon, G. E. (2005). Particle Engineering: A
Formulator’s Perspective. AAPS Arden House Conference.
8.5 CONCLUSIONS Harriman, New York.
13. US Pharmacopeia 27. (2006). US Pharmacopeial convention,
Rockville, MD, pp. 2669–2670.
As timelines become tighter and shortened, it has 14. Mohanmmadi, M.S. & Harnby, N. (1997). Bulk density mod-
become more important than ever to quickly and effi- eling as a means of typifying the microstructure and flow char-
ciently characterize the critical properties of materials acteristics of cohesive powders. Powder Technology 92, 1–8.
that will influence product development and perform- 15. Abdullah, E.C. & Geldart, D. (1999). The use of bulk density
measurements as flowability indicators. Powder Technology
ance. In this chapter, a discussion of those particle, pow- 102, 151–165.
der, and compact properties that are most important 16. Suzuki, M., Sato, H., Hasegawa, M. & Hirota, M. (2001). Effect
in developing solid dosage forms has been discussed. of size distribution on tapping properties of fine powder.
The focus has been on methods that yield important Powder Technology 118, 53–57.
information, yet require small quantities of materials. 17. US Pharmacopeia 27. (2006). US Pharmacopeial convention,
Rockville, MD, pp. 2638–2639.
With a sound understanding of these properties, for- 18. Amidon, G.E. & Houghton, M.E. (1985). Powder flow testing in
mulation development can proceed most efficiently preformulation and formulation development. Pharm. Manuf.
and scientifically with greater success. Tomorrow’s 2(7), 20–31.
formulation and process scientists will require a sound 19. Köhler, T. Influence of Particle Size Distribution on the Flow
understanding of these pharmaceutical material sci- Behaviour of Fine Powders. Particle and Particle Systems
Characterization, 8, 101–104.
ence principles, and must be able to apply them to the 20. Shinohara, K. (2000). Effect of particle shape on angle of internal fric-
design and development of dosage forms in an efficient tion by triaxial compression test. Powder Technology 107, 131–136.
and scientifically rigorous way. The beauty of science is 21. Podczeck, F. (1996). The influence of particle size and shape on
the knowledge and ability it gives us to reliably predict the angle of internal friction and the flow factor of unlubricated
the future. As formulation and process scientists, the and lubricated powders. International Journal of Pharmaceutics
144, 187–194.
future we need to accurately predict is that of a consist- 22. Kachrimanis, K., Karamyan, V. & Malamataris, S. (2003).
ent, reliable, manufacturable product that performs as Artificial neural networks (ANNs) and modeling of powder
expected. flow. International Journal of Pharmaceutics 250, 12–23.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


184 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

23. Moreno-Atanasio, R., Antony, S.J. & Ghadiri, M. (2005). Analyis 45. Fitzpatrick, J.J., Barringer, S.A. & Iqbal, T. (2004). Flow prop-
of flowability of cohesive powders using Distinct Element erty measurement of food powders and sensitivity of Jenike’s
Method. Powder Technology 158, 51–57. hopper design methodology to the measured values. Journal of
24. Tomas, R.J. (2001). Assessment of mechanical properties of cohe- Food Engineering 61, 399–405.
sive particulate solids. Part 2: Powder flow criteria. Particulate 46. Schulze, D. (1994). A new ring shear tester for flowability and
Science and Technology 19, 111–129. time consolidation measurements. Proc. 1st International
25. Carr, R.L. (1965). Classifying flow properties of solids. Chemical Particle Technology Forum, August. Denver/Colorado, USA,
Engineering 72, 69–70. pp. 11–16.
26. Podczeck, F. & Newton, J.M. (1999). Powder filling into hard 47. Jenike, A. W. (1964). Storage and flow of solids. Bulletin 123.
gelatine capsules on a tamp filling machine. International Engineering Experiment Station, University of Utah.
Journal of Pharmaceutics 185, 237–254. 48. Dietmar Schulze. (2002–2004). Ring Shear Tester RST-XS
27. Grey, R.O. & Beddow, J.K. (1969). On the Hausner Ratio and Operating instructions.
its relationship to some properties of metal powders. Powder 49. ASTM International. Standard shear test method for bulk solids
Technology 2(6), 323–326. using the Schulze ring shear tester, D 6773–02.
28. Ho, R., Bagster, D.F. & Crooks, M.J. (1977). Flow studies on 50. ASTM International. Standard test method for shear testing of
directly compressible tablet vehicles. Drug Development and bulk solids using the Jenike shear cell, D 6128–00.
Industrial Pharmacy 3, 475. 51. Hiestand, E. N. & Wells, J. E. (1977). A simplified shear cell
29. Nelson, E. (1955). Measurement of the repose angle of a tab- apparatus and procedure. In: Proceedings of the International
let granulation. Journal of the American Pharmaceutical Powder and Bulk Solids Handling and Process Conference.
Association, Scientific Edition 44, 435. Rosemont, IL, May, p. 244.
30. Armstrong, N.A. & Griffiths, R.V. (1970). The effects of moisture on 52. Schwedes, J. (2000). Testers for measuring flow properties of
the flow properties and compression of phenacetin paracetomol particulate solids. Powder Handling Process 12(4), 337–354.
and dextrose monohydrate. Pharmaceutica acta Helvetiae 45, 692. 53. Kaye, B.H. (1997). Characterizing the flowability of a powder
31. US Pharmacopeia 27. (2006). US Pharmacopeial convention, using the concepts of fractal geometry and chaos theory. Particle
Rockville, MD, pp. 3017–3020. and Particle Systems Characterization 14, 53–66.
32. Khanam, J. & Nanda, A. (2005). Flow of granules through cylin- 54. Hancock, B.C. (2004). Development of a robust procedure for
drical hopper. Powder Technology 150, 30–35. assessing powder flow using a commercial avalanche testing
33. Amidon, G.E. (1995). Physical and mechanical property instrument. Journal of Pharmaceutical and Biomedical Analysis
characterization of powders. In: Physical Characterization of 35, 979–990.
Pharmaceutical Solids, H.G. Brittain, (ed.), Vol. 70. Dekker, New 55. Hickey, A.J. & Concessio, N.M. (1994). Flow properties of
York. pp. 281–319. selected pharmaceutical powders from a vibrating spatula.
34. Grossmann, J. & Tomas, J. (2006). Flow properties of cohesive Particle and Particle Systems Characterization 11, 457–462.
powders tested by a press shear cell. Particulate Science and 56. Rastogi, S. & Klinzing, G.E. (1994). Particle and Particle Systems
Technology 24, 353–367. Characterization 11, 453–456.
35. Schwedes, J. & Schulze, D. (1990). Measurement of flow proper- 57. Doherty, R., Sahajpal, H., Poynter, R. & Lee, Y. (1999). The
ties of bulk solids. Powder Technology 61, 59–68. Journal of Pharmacy and Pharmacology 51, S323.
36. Hiestand, E.N. & Wilcox, C.J. (1968). Some measurements of 58. Boothroyd, E.M., Doherty, R.A., Poynter, R. & Ticehurst, M.D.
friction in simple powder beds. Journal of Pharmaceutical (2000). Comparison of blend flow measured on the Aero-Flow™
Sciences 57, 1421–1427. with tablet weight uniformity. The Journal of Pharmacy and
37. Ho, R., Bagster, D.F. & Crooks, M.J. (1977). Flow studies on Pharmacology 52, 174.
directly compressible tablet vehicles. Drug Development and 59. Boothroyd, E.M., Doherty, R.A., Poynter, R. & Ticehurst, M.
Industrial Pharmacy 3, 475. (2000). The Journal of Pharmacy and Pharmacology 52, 174S.
38. Marchall, K. & Sixsmith, D. (1976). The flow properties of 60. Bhattachar, S.N., Hedden, D.B., Olsofsky, A.M., Qu, X.,
microcrystalline cellulose powders. The Journal of Pharmacy Hsieh, W.Y. & Canter, K.G. (2004). Evaluation of the vibra-
and Pharmacology 28, 770. tory feeder method for assessment of powder flow properties.
39. Fizpatrick, J.J., Barringer, S.A. & Iqbal, T. (2004). Flow property International Journal of Pharmaceutics 269, 385–392.
measurement of food powders and sensitivity of Jenike’s hop- 61. Hiestand, E.N. (2000). Mechanics and Physical Principles for
per design methodology to the measured values. Journal of Powders and Compacts. SSCI Inc, West Lafayette, IN.
Food Engineering 61, 399–405. 62. Hiestand, E.N. (1996). Rationale for and the measurement
40. Schulze, D. “The behavior of powders and bulk solids,” of tableting indices. In: Pharmaceutical Powder Compaction
Fundamentals of Bulk Solid Mechanics, www.dietmar-schulze. Technology, G. Alderborn, & C. Nystrom, (eds), Vol. 71. Marcel
de/grdle1.html. Dekker, Inc, NewYork. pp. 219–244.
41. Juliano, P., Muhunthan, B. & Barbosa-Canovas, G. (2006). Flow 63. Hiestand, E.N., Bane, J.M., et al. (1971). Impact test for hardness
and shear descriptors of preconsolidated food powders. Journal of compressed powder compacts. Journal of Pharmaceutical
of Food Engineering 72, 157–166. Sciences 60(5), 758–763.
42. Podczeck, F. & Mia, Y. (1996). The influence of particle size and 64. Tye, C.K., Sun, C. & Amidon, G.E. (2005). Evaluation of the
shape on the angle of internal friction and the flow factor of effects of tableting speed on the relationships between compac-
unlubricated and lubricated powders. International Journal of tion pressure, tablet tensile strength, and tablet solid fraction.
Pharmaceutics 144(2), 187–194. 29 November. Journal of Pharmaceutical Sciences 94(3), 465–472.
43. Podczeck, F. & Newton, J.M. (2000). Powder and capsule filling 65. Rees, J.E. & Rue, P.J. (1978). Time-dependent deformation of
properties of lubricated granulated cellulose powder. European some direct compression excipients. The Journal of Pharmacy
Journal of Pharmaceutics and Biopharmaceutics 50, 373–377. and Pharmacology 30, 601–607.
44. Nyqvist, H. (1982). Prediction of weight variation in tablet pro- 66. David, S.T. & Augsburger, L.L. (1977). Plastic flow during com-
duction from shear cell measurements. Acta pharmaceutica pression of directly compressible fillers and its effect on tablet
Suecica 19, 413–420. strength. Journal of Pharmaceutical Sciences 66(2), 155–159.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


8.5 CONCLUSIONS 185

67. Roberts, R.J. & Rowe, R.C. (1986). The effect of the relationship 86. Mullarney, M.P., Hancock, B.C. & Narajan, P. (2004). Improving
between punch velocity and particle size on the compaction the prediction of exceptionally poor tableting performance:
behaviour of materials with varying deformation mechanisms. an investigation of Hiestand’s “special case”. Journal of
The Journal of Pharmacy and Pharmacology 38, 567–571. Pharmaceutical Sciences 93(8), 2017–2021.
68. Ishino, R., Yoshino, H., Hirakawa, Y. & Noda, K. (1990). Influence 87. Wu, C.Y., Best, S.M., Bentham, A.C., Hancock, B.C. & Bonfield, W.
of tabletting speed on compactibility and compressibility of two (2005). A simple predictive model for the tensile strength of
direct compressible powders under high speed compression. binary tablets. European Journal of Pharmaceutical Sciences
Chemical & Pharmaceutical Bulletin 38(7), 1987–1992. 25(2–3), 331–336.
69. Ruegger, C.E. & Celik, M. (2000). The effect of compression and 88. Wu, C.Y., Ruddy, O.M., Bentham, A.C., Hancock, B.C.,
decompression speed on the mechanical strength of compacts. Best, S.M. & Elliott, J.A. (2005). Modelling the mechanical
Pharmaceutical Development and Technology 5(4), 485–494. behaviour of pharmaceutical powders during compaction.
70. Armstrong, N.A. & Palfrey, L.P. (1989). The effect of machine Powder Technology 152(1–3), 107–117.
speed on the consolidation of four directly compressible tablet 89. Hancock, B.C., Carlson, G.T., Ladipo, D.D., Langdon, B.
diluents. The Journal of Pharmacy and Pharmacology 41, 149–151. A. & Mullarney, M.P. (2002). Comparison of the mechanical
71. Hancock, B.C., Colvin, J.T., Mullarney, M.P. & Zinchuk, A.V. properties of the crystalline and amorphous forms of a drug
(2003). The relative densities of pharmaceutical powders, substance. International Journal of Pharmaceutics 241(1),
blends, dry granulations, and immediate-release tablets. 73–85.
Pharmaceutical Technology 27(4), 64–80. 90. Amidon, G.E. & Houghton, M.E. (1995). The effect of moisture
72. Maarschalk, K.V., Vromans, H., Groenendijk, W., Bolhuis, G.K. & on the mechanical and powder flow properties of microcrys-
Lerk, C.F. (1997). Effect of water on deformation and bond- talline cellulose. Pharmaceutical Research 12(6), 923–929.
ing of pregelatinized starch compacts. European Journal of 91. Amidon, G. E. (2006). Data driven formulation development
Pharmaceutics and Biopharmaceutics 44(3), 253–260. using material sparing methods. Garnet Peck Symposium,
73. (1996). Pharmaceutical Powder Compaction Technology, G. Alderborn & Purdue University, September 20–21. West Lafayette, IN.
C. Nystrom (eds), Vol 71. Marcel Dekker, Inc., NewYork. 92. Amidon, G. E. (2002). Mechanical property characterization
74. Hiestand, E., Wells, J.E., Peot, C.B. & Ochs, J.F. (1977). Physical of API, excipients and formulations during development.
processes of tableting. Journal of Pharmaceutical Sciences 66(4), AAPS Pharmaceutics and Drug Delivery Conference, April.
510–519. Washington, DC.
75. Hiestand, E.N., Bane, J.M. & E.P., (1971). Strzelinski impact 93. Leuenberger, H. & Leu, R. (1992). Formation of a tablet: A site
test for hardness of compressed powder compacts. Journal of and bond percolation phenomenon. Journal of Pharmaceutical
Pharmaceutical Sciences 60(5), 758–763. Sciences 81(10), 976–982.
76. Hiestand, E.N. & Smith, D.P. (1984). Indices of tableting per- 94. Fell, J.T. (1996). Compaction Properties of binary Mixtures. In:
formance. Powder Technology 38, 145–159. Pharmaceutical Powder Compaction Technology, G. Alderborn, &
77. Hiestand, E.N. & Smith, D.P. (1991). Tablet Bond I. A theoretical C. Nystrom, (eds), Vol. 71. Marcel Dekker, Inc, NewYork. pp.
model. International Journal of Pharmaceutics, 67, 217–229. 501–516.
78. Hiestand, E.N. & Smith, D.P. (1991). Tablet Bond II. Experimental 95. Wurster, D.E., Majuru, S. & Oh, E. (1999). Prediction of the
check of model. International Journal of Pharmaceutics, 67, Hiestand bonding indices of binary powder mixtures from sin-
231–246. gle-component bonding indices. Pharmaceutical Development
79. Fell, J.T. & Newton, J.M. (1970). Determination of tablet strength and Technology 49(1), 65–70.
by the diametral-compression test. Journal of Pharmaceutical 96. Amidon, G. E. (2006). Mechanical property characterization
Sciences 59(5), 688–691. of pharmaceutical mixtures. Department of Materials Science,
80. He, X., Secreast, P.J. & Amidon, G.E. (2007). Mechanistic August 2. University of Cambridge, UK.
study of the effect of roller compaction and lubricant on tablet 97. Lammens, R.F. & Fedin, V.P. (1978). Modern press instrumen-
mechanical strength. Journal of Pharmaceutical Sciences 96(5), tation Journal of Medicinal and Pharmaceutical Chemistry
1342–1355. 12, 263.
81. Mullarney, M.P., et al. (2003). The powder flow and compact 98. Paronen, P. & Ilkka, J. (1996). Porosity-Pressure Functions. In:
mechanical properties of sucrose and three high-intensity Pharmaceutical Powder Compaction Technology, G. Alderborn, &
sweeteners used in chewable tablets. International Journal of C. Nystrom, (eds), Vol. 71. Marcel Dekker, Inc, NewYork. .
Pharmaceutics 257(12), 227–236. 99. Huxley Bertram Engineering Ltd. Cambridge, UK, http://
82. Boldyreva, E.V., Dmitriev, V. & Hancock, B.C. (2006). Effect of www.powder-compaction.com/compaction-simulators.htm
pressure up to 5.5 GPa on dry powder samples of chlorpropa- 100. Metropolitan Computing Corp., http://www.mcc-online.com/
mide form-A. International Journal of Pharmaceutics 327(1–2), presster.htm
51–57. 101. Heckel, R.W. (1961). Density–pressure relationships in powder
83. Hancock, B.C., et al. (2001). The powder flow and compact compaction. Trans Metallurgical Society of AIME 221, 671.
mechanical properties of two recently developed matrix-form- 102. Cooper, A.R & Eaton, L.E. (1962). Compaction behavior of
ing polymers. The Journal of Pharmacy and Pharmacology several ceramic powders. J Am Ceram Soc. 45, 97–101.
53(9), 1193–1199. 103. Nelson, E., Busse, L.W. & Higuchi, T. (1955). Journal of the
84. Hancock, B.C., Dalton, C.R. & Clas, S. (2001). Micro-scale American Pharmaceutical Association Scientific Edition
measurement of the mechanical properties of compressed 44, 223.
pharmaceutical powders 2: The dynamic moduli of microcrys- 104. Hersey, J.A. & Rees, J. (1971). Deformation of particles during
talline cellulose. International Journal of Pharmaceutics 228(1– briquetting. Nature PS 230, 96.
2), 139–145. 105. York, P. & Pilpel, N. (1973). J. Pharm. Pharmacol. 25, 1.
85. Wu, C.Y., Best, S.M., Bentham, A.C., Hancock, B.C. & Bonfield, W. 106. Maarschalk, K.V., Zuurman, K., Vromans, H., Bolhuis, G.K. &
(2006). Predicting the tensile strength of compacted multi-com- Lerk, C.F. (1996). Porosity expansion of tablets as a result of
ponent mixtures of pharmaceutical powders. Pharmaceutical bonding and deformation of particulate solids. International
Research 23(8), 1898–1905. Journal of Pharmaceutics 140(2), 185–193.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS


186 8. PARTICLE, POWDER, AND COMPACT CHARACTERIZATION

107. Joiris, E., Di Martino, P., Berneron, C., Guyot-Hermann, A.M. & 110. Hersey, J.A. & Rees, J. (1971). Deformation of particles during
Guyot, J.C. (1998). Compression behavior of orthorhombic briqueting. Nature (PhysSci) 230, 96.
paracetamol. Pharmaceutical Research 15(7), 1122–1130. 111. Celik, M., Ong, J.T.H., Chowhan, Z.T. & Samuel, G.J. (1996).
108. Duberg, M. & Nystrom, C. (1982). Studies on direct compression Compaction simulator studies of a new drug substance:
of tablets. VI. Evaluation of methods for the estimation of Effect of particle size and shape, and its binary mixtures with
particle fragmentation during compression. Acta Pharmaceutica microcrystalline cellulose. Pharmaceutical Development and
Suecica 19, 421–436. Technology 1(2), 119–126.
109. Heckel, R.W. (1961). Density–pressure relationships in powder
compaction. Trans Metall Soc AIME 221, 671–675.

I. THEORIES AND TECHNIQUES IN THE CHARACTERIZATION OF DRUG SUBSTANCES AND EXCIPIENTS

You might also like