Download as pdf or txt
Download as pdf or txt
You are on page 1of 33

Sponsored by

Optimizing Pharmaceutical Formulations:


The Role of Physicochemical Analysis
Contents
3 Introduction

4 Answering the Challenge of Setting Effective Particle Size Specifications


During Pharmaceutical Product Development

12 Discriminating Between Polymorphs of Acetaminophen Using


Morphologically-Directed Raman Spectroscopy

16 Laboratory PXRD Systems Can Address Pharmaceutical R&D Challenges

20 An Interview with Andrew L. Theophilus, R&D Manager -


Physical Properties, Chemo R&D

24 Comparing Your Material’s Structure to Literature and Patent


PXRD Patterns

26 Automated Morphological Imaging: Deeper Insight into your


Pharmaceutical Formulation or Product

30 An Interview with Paul Kippax, Pharmaceutical Sector Director -


Malvern Panalytical

2
Introduction
Developing drug products is a progressively competitive, expensive and high-risk venture that
when successful, can bring considerable humanitarian and financial reward. At all stages of the
pharmaceutical development process, developers need to increase efficiency while maintaining
quality and safety – beginning with drug discovery, through chemical and formulation
development, to manufacturing and QC.

In recent decades, formulation development and optimization has become an increasingly


systematic, knowledge-driven process. Developing a successful formulation relies on a detailed
scientific understanding of the behavior of individual components, and of the interaction and
combined effect of these constituents in the candidate formulation.

Malvern Panalytical’s unique range of physicochemical analysis techniques can be used to


accelerate the development of pharmaceutical formulations, enabling critical material attributes
such as particle size, shape, structure and composition to be measured, aiding reference listed
drug (RLD) deformulation, helping establish bioequivalence in vitro, and accelerating formulation
development efficiency for both innovator and generic products.

This eBook explores several ways in which Malvern Panalytical’s technologies and expertise can
assist pharmaceutical scientists in their quest to develop high quality, safe and effective products,
and accelerate their speed to market.

3
Answering the Challenge
of Setting Effective
Particle Size Specifications
During Pharmaceutical
Product Development
Setting appropriate product and processing spec-
Introduction
ifications is fundamental to QbD. Clinical perfor-
mance is a function of key variables controlled by
Setting meaningful and realistic specifications is an
and during the manufacturing process. Identifying
essential element of Quality by Design (QbD). Well-
these Critical Quality Attributes (CQAs) and con-
defined specifications control product performance trolling them within clearly-defined ranges delivers
since they derive from correlations between clini- acceptable product quality and the required in vivo
cal behavior and the variables measured routinely behavior. Getting specifications right is therefore es-
during processing and for QC. This paper examines sential, but demanding. An overly-tight specification
the process of setting specifications, taking as an ex- will make manufacturing more difficult than it needs
ample particle size, a critically important parameter to be, compromising production economics, yet one
for many pharmaceutical formulations. that is too loose may result in product inconsisten-
cies that impact efficacy.
Multiple pressures on the pharmaceutical industry
Here we consider the process of specification setting
and the regulatory bodies that control it are driving
in some detail, using particle size as an example pa-
the shift towards a risk-based approach to produc-
rameter. Particle size influences the key performance
tion. Increasingly, regulators are focusing scarce re- characteristics of many of the suspensions, inhaled
source on those less well-understood products and powders and tablets widely used in drug delivery.
processes with significant potential for harm, re-
sponding more flexibly to manufacturers who can Establishing the Need for
demonstrate sound understanding and control. a Specification
QbD is a key part of this change in emphasis, pro-
moting development of the knowledge through FDA guidance enshrined in ICH topic Q6A states
which manufacturers will prove competence. that, ‘specifications are critical quality standards that

4
are proposed and justified by the manufacturer and • Dissolution, solubility or bioavailability
approved by regulatory authorities as conditions of • Processability
approval.’ Because specifications define product ac-
ceptability, the requirement for them is based on the • Stability
existence of a link between a variable and some as- • Product content uniformity
pect of performance. Within the framework of QbD, The dependence of dissolution behavior on particle
such correlations establish a parameter as a CQA, the size, or more accurately surface area, is widely recog-
control of which assures product quality. The regula- nized. For solid dosage forms, dissolution behavior
tory guidance suggests these might include: (including overall solubility), influences in vivo re-
• Physicochemical properties such as pH, lease and is often tightly controlled to obtain the re-
refractive index, melting point quired delivery profile. Finer powders dissolve more
• Particle size rapidly, and those with a narrow size distribution all
dissolve at a similar rate, which may or may not be
• Polymorphic form/amorphous content
the design intent. For OINDPs (orally inhaled and
• Chiral identity nasal drug products), the link with bioavailability is
• Water content even more direct because incorrectly sized particles
• Inorganic impurity levels will not deposit within the desired part of the respi-
• Microbial content ratory system, thus failing to enter the body by the
prescribed route.
Clearly if a variable has no influence on product qual-
ity there is no need to specify values for it; equally, Processability is important because it tends to cor-
where there is a correlation, any associated specifica- relate with product consistency and quality, with er-
tion must provide adequate control. The best way to ratic or poorly controlled processes producing a low
define the specification will depend on the mecha- quality output. The aim of QbD is to develop a man-
nism by which the variable influences behavior; opti- ufacturing process on the basis of sound science that
mal definition requires detailed understanding. QbD will deliver long-term optimal manufacture, reducing
emphasizes the need for relevant information at the the reliance on final QC testing. Particle size relates to
early stages of a project, and the importance of de- parameters such as powder flowability, segregation
veloping an adequate knowledge base. The pharma- behavior and, in the case of tableting processes, com-
copoeial standards, development data, test data from pressibility. As such, well-defined size specifications
pre-clinical and clinical studies, and from stability tri- for in-process material or intermediates may provide
als are all good sources of information. a means of reducing process variability.

Focusing on Particle Size Stability issues arise because the forces acting on
particles are a function of their size. Smaller parti-
ICH topic Q6A contains a decision tree that deter- cles are more likely to cluster or agglomerate be-
mines the need for a particle size specification, rec- cause inter-particle forces are relatively high, while
ommending them for both solid dosage forms and with suspensions the propensity to settle increas-
liquids containing undissolved drug suspensions es with size because of the effects of gravitational
when particle size is critical to any of the following: forces. If settling does occur, then the chances of

5
consistent dosing are reduced - unless the active product performance. Correlations with, for exam-
ingredient is successfully resuspended before use. ple, dissolution profile, content uniformity, suspen-
Stability must be considered during the develop- sion stability, and/or mouth feel, form the basis for
ment of particle size measurement methods, to specification development.
ensure that the state of dispersion during analysis
reflects that of the material. Measuring Particle Size
Finally, particle size can influence blend uniformity
Distributions: Laser Diffraction
and, as a result, the dose content uniformity asso-
Before considering how particle size specification
ciated with a product. For example, Figure 1 shows can be set, it is worth reviewing the methods by
a near-infrared (NIR) chemical image of a recalled which particle size analysis can be carried out. There
tablet with the active ingredient-rich domains are now a range of particle size analysis tools which
picked out in red. These areas are much larger on are available to support pharmaceutical develop-
the right side of the tablet than the left, showing ment. Selection of the most appropriate technique
that composition is not uniform. The reason giv- is, in itself, an important consideration, with param-
en for the recall of these tablets was that, ‘due to eters such as the precision and accuracy of measure-
larger particle size some of the unit doses may not ment, the time for analysis, ease of method develop-
meet potency specifications.’ Here then is a direct ment and transfer, and system cost being possible
example of poor particle size control resulting in a selection criteria.
sub-standard product. Development of a particle
size specification may provide a means of control. One of the most widely used techniques for parti-
cle size analysis within the pharmaceutical industry
In summary, particle size specifications are often is laser diffraction. Laser diffraction is a particle size
required for solid dosage forms or suspensions be- measurement technique with broad applicability,
cause particle size influences so many aspects of suitable for both wet and dry systems with particles
in the size range 0.1 to 3500 microns. A particularly
attractive feature of the method is that it is an ide-
al Process Analytical Technology (PAT), so specifica-
tions developed in the laboratory can be transferred
into production, and if the right equipment is used.

How are Size Distributions


Represented?
Using laser diffraction as an example, the first issue
which should be understood in setting a specifica-
tion relates to how the particle size distribution is
represented. As an ensemble sizing method, laser
Figure 1. NIR image of a tablet showing diffraction produces volume based distributions.
heterogeneous distribution of the active ingredient This means that data are generated simultaneously

6
for the whole sample (ensemble) rather than for in- based on the entire distribution, but instead nor-
dividual particles, and are presented in terms of the mally consider specific distribution parameters or
volume of material in a given size fraction. An alter- statistics. Choosing the most relevant parameter
native, used by imaging and microscopy methods and making a judgement as to whether a single fig-
for example, is to display data on a number basis (i.e. ure is adequate or whether multiple descriptors of
show the number of particles in each size fraction). distribution are necessary are important steps in the
Both approaches are equally valid, but the results specification setting process.
produced are very different (see Figure 2). Figure 3 shows an example size distribution, in this
case a mixture of two components, which has been
measured using laser diffraction. Using this, we can
consider which statistics relate to different regions
of the distribution.

Figure 2. Comparing volume and number


based distributions

The number and volume distributions shown in Figure 3. Example size distribution, showing possible
Figure 4, clearly an extreme case, reflect different parameters which could be used in a product specification
attributes of the sample. The volume distribution
The median is the particle size in the middle of the
highlights those size fractions in which most of the
distribution. In the case of laser diffraction, this is
mass or volume lies, while the number distribution
referred to as the Dv50, indicating that 50% of the
captures the presence of fines. Which is the better
measure depends on the application; comparisons of volume of particles are larger and 50% smaller than
number and volume-based distributions are feasible this value (the notation ‘v’ shows that this param-
but best avoided if possible, because of the impact of eter relates to a volume distribution). For the sin-
measurement errors. A better approach is to consider gle Gaussian distribution, median and mode (the
this issue during analytical technique selection. most common particle size) are identical; however
for the bimodal distribution such as the one shown
Defining Particle Size here, the mode reveals nothing about the smaller
peak. The median value on the other hand does re-
Although the measurement of particle size distribu- flect the presence of fines and is located between
tions is extremely useful, specifications are seldom the two populations.

7
A similar notation can be used to describe other
The Importance of Selecting an
points within the size distribution. So, the Dv10 re-
lates to the particle size below which 10% of the vol-
Appropriate Mean
ume of material exists, and is therefore positioned
Consider a particle population consisting of ten par-
towards the fine end of the distribution. In contrast,
ticles of unit diameter and one with a diameter of
the Dv90 is associated with the coarse end of the dis-
100 units. The number mean, or arithmetic average,
tribution. The Dv100 is also shown, and might seem
diameter or D[1,0] for this group of particles is calcu-
a logical choice if the aim is to avoid particles above
lated as follows:
a certain size. However, using Dv100 makes the anal-
ysis sensitive to the presence of just one large par- D[1,0] = (10*1 + 1*100) / (10 + 1) = 10 units
ticle, massively increasing the importance of repre-
sentative sampling. With modern instrumentation, This confirms that the D[1,0] is weighted towards the
sampling is often the largest source of error, so this fines. If the large particle breaks into two, each with
approach is not recommended. Using Dv90 or even a diameter of 79.37 units (thereby conserving mass)
Dv95 can provide a more robust parameter for as- then the D[1,0] for the population becomes:
sessing the coarse fraction. D[1,0] = (10*1 + 2*79.37) / (10 + 2) = 14.06 units
The other term commonly used for the description
This is a 40.6% increase in size, and as such clearly
of statistical data is mean or average. However, in
fails to reflect particle attrition.
selecting a mean size parameter, it is important to
understand how the mean is calculated. The mean On the other hand, we could consider the D[4,3].
may be ‘weighted’ according to different particle Here, the proportion of particles at each size is
parameters. Typical parameters include the num- weighted according to their volume when calcu-
ber or arithmetic mean diameter, the D[1,0], surface lating the mean size. So for the first population, the
area moment mean, the D[3,2], or volume moment D[4,3] is:
mean, the D[4,3]. Clearly the different weightings for
each of these means will yield different results, as D[4,3] = (10*1*13 + 1 * 100 *1003) / (10 * 13 + 1 *
shown in Figure 3. 1003) ≈ 100 units

The moment means are more commonly used for This confirms that the D[4,3] is weighted towards the
specifications: the D[3,2] shows a greater sensitivity coarse material. The break-up of the large particle
to fines content, as the fine particles have a high into two results in the following D[4,3]:
specific surface area, whereas the D[4,3] is partic- D[4,3] = (10*1*13 + 2 * 79.37 *79.373) / (10*13 +
ularly useful for the detection of larger particles,
2*79.373) ≈ 79.37 units
as these contain the greatest volume of material.
Where the mean is the basis for a specification, se- This moment of distribution mean is clearly able to de-
lecting an appropriately sensitive measure is there- tect the change caused by particle break-up, although
fore essential. it shows little sensitivity to the fines population.

8
Selecting an Appropriate Parameter
Measuring how sensitive different particle size pa-
rameters are to changes in a sample is a good way
of determining the best basis for a specification.
Figure 4 shows particle size distributions, mea-
sured for the blending of two excipient grades,
generated using laser diffraction (Mastersizer 2000,
Malvern Panalytical).

Figure 5. The impact of the percentage of fine material on


different size descriptors

For this example it is obvious that Dv10 and D[3,2]


can only track changes in the particle size at low
fines content (<10%), whereas Dv90 only shows
sensitivity at high fines contents (>40%). In contrast,
the Dv50 and D[4,3] show a linear change as the
percentage of fine material increases, and therefore
provide a means of differentiating between blends
across the range tested here. These parameters may
Figure 4. Investigating the ability of a laser diffraction therefore be more appropriate for a specification, al-
analyzer (Mastersizer 2000) to detect fine and coarse
though this does depend on the range of possible
particles in a pharmaceutical blend
blends which could occur in production. If the fines
Here the aim was to assess the ability of the instru- content is never greater than 10%, the Dv10 or D[3,2]
ment to detect small changes in the fines content. may provide a more sensitive measure of change, as
Detecting the relative proportion of fines and coarse the rate of change of these parameters is greatest
material in a blend is a common application, since below 10% fines.
particle size distribution often has an influence on
important properties such as flowability. In the for- Setting Tolerances
mulation of dry powder inhalers, for example, the
inclusion of fine excipient particles blended with Techniques such as laser diffraction offer excellent
coarser carrier particles is an established way of repeatability, reproducibility and robustness, pro-
enhancing drug delivery, because it improves the ducing good quality data with minimal manual in-
aerosolization process. Figure 5 shows how various put or expertise. High repeatability means that the
size descriptors change with increasing fine material same sample, run on the same system, produces
content in this case. very similar results, so it is solely dependent on the

9
instrument itself and on reproducible sample dis- must be 10 microns size or less would be subject to
persion. Reproducibility is a more demanding pa- a measurement variability of +/-14%.
rameter that quantifies the variability introduced by
a change of operator, sample, time and instrument; This analysis highlights a fairly obvious point: as
sampling methodology consequently becomes im- measurement variability increases, confidence in
portant. Measurement variability informs the final the reported result should reduce. Although this
step of specification setting: the determination of concept is readily appreciated, it is not always prop-
acceptable tolerances. erly accounted for in specifications. Here is a spec-
ification for a tableting blend [Evolutions in Direct
Setting appropriate tolerances for a specification Compression, Douglas McCormick, Pharmaceutical
requires an appreciation of measurement errors as Technology, April 2005. Pg 52-62]:
well as an understanding of the correlation between
the variable and product performance. Figure 6 Dv10 > 30 μm
shows some particle size distribution data, the red D[4,3] > 80 μm
line being a typical reading, the yellow and orange
lines quantifying reproducibility, the range of vari- Dv90 < 1000 μm
ability associated with the reading.
The tolerances above do not take into account any
If the specification for this product is a Dv50 of 10 error introduced by measurement, but rather re-
microns, then the plot shows that the measurement flect the ideal material for optimal production. Dv10
variability will be +/- 5%. However, it would be wrong should be greater than 30 microns, D[4,3] greater
to assume that the same tolerance can also be used than 80 microns, and the Dv90 should be less than
for the percentage less than 10 microns. Due to the 1000 microns. USP <429> [General Chapter <429>
slope of the undersize distribution curve, a specifica- (2008), “Light Diffraction Measurements of Particle
tion requesting that 50% of the volume of material Size,” United States Pharmacopeia. www.usp.org]
provides acceptable reproducibility figures for par-
ticle size measurement, suggesting that the Dv50
and other central measures should be within 10%,
and noncentral values such as Dv10 and Dv90
should be within 15%. If the analytical technique
used delivers this performance, what does that
mean for the specification?

If the USP <429> reproducibility limits are applied, a


Dv10 measurement of 30 microns could be recorded
for a sample with a Dv10 of up to 34.5 microns (15%
error). If the aim is to ensure that the material actual-
ly has a Dv10 greater than 30 microns, the specifica-
tion will need to be changed accordingly. Applying
Figure 6. Highlighting how accuracy is affected by the the same approach for each parameter produces the
wording or basis of a specification following measurement specification which needs

10
to be followed if the manufacturing specification within the industry. Recognizing the parameters
above is to be met: that different particle sizing techniques deliver and
the relevance of alternative size descriptors is crucial
Dv10 > 34.5 μm
for the development of a relevant specification, as is
D[4,3] > 88 μm the refinement of tolerances in line with the accura-
cy of the method.
Dv90 < 850 μm
Laser diffraction is an ensemble particle sizing tech-
So, tolerances should be reduced to incorporate the
nique that delivers volume-based distributions. It is
errors introduced by the measurement technique,
suitable for many applications within the sector and
with less accurate methods requiring tighter speci-
is particularly sensitive for the detection of over-
fications. This provides impetus for the procurement
sized material. Systems such as the Mastersizer 2000
of instruments with high reproducibility and the de-
velopment of robust methodologies. deliver highly repeatable results (+/-1%) reducing
the need to tighten tolerances to account for mea-
In Conclusion surement inaccuracy. Furthermore, because laser
diffraction is well-established as a process analytical
Setting specifications on the basis of product under- technique, a specification developed in the labora-
standing is a key element of QbD and essential for tory can be transferred during scale-up. The success-
effective quality control. For many pharmaceuticals, ful transfer of specifications that are as broad as pos-
particle size is a CQA – a variable that must be tightly sible, within the constraints of meeting performance
controlled because it directly impacts performance targets, is a route to optimized manufacture and re-
– so, specification and measurement is routine duced cost.

11
Discriminating Between
Polymorphs of Acetaminophen
Using Morphologically-Directed
Raman Spectroscopy
morphological and spectroscopic results, where
Introduction
morphologically-distinct particle groups are found
Interest in studying mixed systems of pharmaceuti- to share common spectra, and vice versa. The ability
cal relevance is not a new concept. From intrinsic of MDRS to target particles of distinct morphology
polymorphism to complex final products, the chal- for Raman investigation provides added sensitivity
lenges encountered by modern analytical scientists to the measurement, as spectra for particles that are
have multiplied both in number and in complexity. not of interest are not measured. Likewise, know-
Established techniques are often unable to provide ing that a group of particles with similar spectra may
both separation and identification of species in a be described by a common morphology allows the
mixed system, as they are used to probe chemistry analyst to assess particulate quantity in the absence
(bulk spectroscopy), or individual particle character- of measuring a spectrum for each particle in the
istics (microscopy and SEM). Combining techniques morphologically-similar group. Such correlations
capable of analyzing micron-scale particulates with between particle morphology and chemical identity
absolute chemical specificity provides a new win- allow MDRS to provide an improvement in efficiency
dow into describing the complicated mixtures com- over manual methods.
mon in today’s competitive pharmaceutical market.
Materials and Methods
Malvern Panalytical’s Morphologi® ID provides just
such a technology combination, uniting automated A polymorphic mixture of common pain-relief
microscopy and spectroscopy in an integrated pack- medication was investigated using a Morphologi
age. This partnering of techniques has been termed ID instrument to characterize samples in terms
Morphologically-Directed Raman Spectroscopy of particle size, shape, and chemical identity. The
(MDRS®). Raman spectroscopy is sensitive to the polymorphic mixture consisted of the Type I and
unique polymorphic forms of a substance, and Type II polymorphs of acetaminophen (also known
can be used to investigate single particles to assist as paracetamol). Acetaminophen in its Type I form
in determining the long-term stability of a prod- (Fisher (98%, AC102332500)) was recrystallized by
uct. Correlations may be established between slowly cooling a saturated solution in water at 60°C.1

12
The resulting material was recovered and dried at
Results
75°C for eight hours, at which point half of the sam-
ple (Type I) was removed and the other half heated Over 70,000 individual particles were detected for
to 170°C to effect the Type I to Type II polymorphic the 9 (Type I):1 (Type II) acetaminophen mixture.
conversion.2 A volumetric fraction consisting of 9
The sample was polydisperse, with some particles
parts Type I to 1 part Type II by weight was milled
greater than 400 μm detected. Examples of the par-
for 1 minute in a hand mill. 3 mm3 of the resulting
ticles analyzed and their final size distribution are
mixed powder was placed into the dry dispersion
shown in Figure 1.
accessory for the Morphologi ID instrument and dis-
persed at a pressure of 4 bar onto a quartz plate. The morphological results show that the hand mill
A 20 mm diameter area was scanned at 5x magnifi- produces a broad distribution of particle sizes. It
cation to detect acetaminophen particles, and 1375 is also apparent that a few large particles (greater
particles were selected for Raman measurement than 200 μm) are still present in the final disper-
based on the following size categories: greater than sion. The size distribution result is used to classify
50 μm, 25 μm to 50 μm, and 10 to 25 μm. the particles based on size (10 μm to 25 μm, 25 μm
to 50 μm, and greater than 50 μm) for Raman in-
Reference spectra for Raman analysis were obtained vestigation. Particles below 10 μm were excluded
from pure materials. Spectra were collected with a from Raman analysis.
785 nm laser (20 mW) illuminating a 3 μm spot size
at 50x magnification, and covering a range from The results of correlation against the library of pure
150 cm-1 to 1850 cm-1 at 6 cm-1 resolution. polymorph spectra revealed 1,220 Type I particles

Figure 1. Representative particle images from the polymorph acetaminophen mixture (left panel) and the size distribution
(volume weighted, right panel).

13
and 144 Type II particles, out of 1,375 particles tar- mixture spectra identified for a subset of particles
geted, corresponding to 10.5% Type II polymorph. are most likely due to the formation of aggregates
The pure reference spectra and examples of particle during dispersion.
spectra are shown in Figure 2. The correlation was
constrained to contain only the 1200 cm-1 to 1260 Once the spectroscopic data is available, the mor-
cm-1 and 1540 cm-1 to 1590 cm-1 regions (indicated phologies of the two species can be reviewed to
by the green areas in Figure 2) where the two spe- determine if it is possible to segregate similar look-
cies are readily distinguishable. A correlation score ing materials based solely on their morphologies. In
of greater than 0.85 was required for a positive iden- the present example, results confirm that the Type I
tification as either Type I or Type II. Mixture spectra particles have a higher Intensity Mean than the Type
(correlation to both Type I and Type II references
II particles, meaning that Type I particles are more
with scores greater than 0.85) were detected for 11
transparent, as shown in Figure 3. However, while a
particles, an example is also shown in Figure 2.
clear difference in the transparency of the two parti-
The agreement between the recorded and library cle types exists, it should be noted that there is some
spectra is outstanding, and allows for simple dis- overlap between the Intensity Mean distributions
crimination between the two polymorph types which suggests that it is not a definitive morpholog-
within the confines of the correlation analysis. The ical separator. No other morphological descriptors
indicated a significant difference between the par-
ticle types, most likely due to loss of morphological
information during the milling process. This situa-
tion emphasizes the importance of having a ‘referee’
technique, such as Raman spectroscopy, available to
discriminate between different chemical species.

Figure 2. Raman spectra collected from example


particulates for acetaminophen Type I (blue),
acetaminophen Type II (red), and a mixture spectrum
(black). The Type I and Type II spectra are superimposed Figure 3. Intensity Mean distribution for the Type I and
over reference spectra used to create the library (black Type II particles as determined by the Raman spectroscopic
dotted). The portions of the spectrum in green are those results. Insets display example images of particles from
used for correlation to the reference library. each group.

14
directly. Spectra are collected for single particles,
Discussion
which allow at least an order of magnitude increase
Polymorphic conversions are considered an ongo- in detection sensitivity. Data collection can be con-
ing problem in the pharmaceutical industry. Process ducted for any number of particulates desired, and
conditions, improper storage and simply time can unique spectra are easily identified. As mentioned
cause the transition from a stable, efficacious form previously, the morphological characteristics of
of a drug substance into an unwanted structural particulates can also be used to further refine the
analog. Thus, establishing the presence of multi- selectivity of the method. If different polymorphic
ple polymorphic forms is of great interest to both forms have distinct morphology, the method can
innovator and generic pharmaceutical companies be modified to investigate only those that fit a par-
alike. Vibrational spectroscopy has found wide use ticular morphological profile. These strengths posi-
as a tool with the requisite sensitivity to establish tion MDRS as a technique that can handle the most
the presence of multiple polymorphic forms in a difficult polymorphic mixtures with unparalleled
mixed system. Unfortunately, commonly-available sensitivity and flexibility, providing a new tool for
bulk spectroscopic techniques lack the sensitivity to root cause analysis, product quality and formulation
detect the low levels of polymorphic contaminants development.
that may be present in final products. Additionally,
bulk techniques provide only an overview of the ma- References
terial that is present in the sample area and have no
ability to partition the sample into individual parti- 1. Lee T., Kuo C.S., and Chen Y.H. Oct. 2, 2006. Solubility,
cles to provide specific characterization. Polymorphism, Crystallinity, and Crystal Habit of Acetaminophen
and Ibuprofen by Initial Solvent Screening. Pharmaceutical

Conclusion Technology.

2. Di Martino P., Conflant P., Drache M., Huvenne J.-P., and Guyot-
MDRS provides clear advantages over traditional Hermann A.-M. 1997. Preparation and Physical Characterization of
methods of vibrational spectroscopy and is well-suit- Forms II and III of Paracetamol. Journal of Thermal Analysis (48):
ed to address low-level polymorphic contamination 447-458.

15
Laboratory PXRD Systems
Can Address Pharmaceutical
R&D Challenges
Detection of trace polymorphs, determination of optics achieve excellent LOD by suppressing the
amorphous content, and the study of the stability of continuous radiation (Bremsstrahlung) and there-
active pharmaceutical ingredients (APIs) subjected to fore improving the signal to noise ratio in the data.
heat and humidity are some of the many applications One such optic for greatly improved limits of de-
where a modern powder X-ray diffraction (PXRD) sys- tection is the Malvern Panalytical BraggBrentanoHD
tem can add value to drug development. Advances (BBHD) optic.
in beam conditioning and detector technology have
led to greatly improved detection limits, resolution The following example shows part of the diffrac-
and range of applications now possible. Today’s mod- tion pattern for an Indomethacin (IMC) mixture
ern, laboratory-based PXRD instrumentation and ad- of two crystalline polymorphs in the excipient.
vanced software can greatly assist pharmaceutical The mixture was made with 0.25% alpha-Indo-
scientists in their quest to develop high quality, sta- methacin, 4.75% gamma-Indomethacin and two
ble products and accelerate their speed to market. excipients (98.5% alpha-lactose monohydrate,
All applications described and many more can be 1.5% Magnesium stearate). Figure 1a. shows the
performed on a single PXRD instrument, such as the alpha-Indomethacin peak measured with a BBHD
Malvern Panalytical Empyrean. optic with a significant improvement of the LOD
to 0.12%, as compared to standard slit optics in
Detecting the Presence of Trace Figure 1b. BBHD offers further benefits to phar-
Polymorphs and Impurities maceutical research, with its improved low angle
performance as compared to slit optics, and the
The detection of trace polymorph phases is one of suitability to be used for several applications be-
the most important applications for PXRD in the yond powder diffraction, including small angle
pharmaceutical industry. While classical PXRD con- X-ray scattering (SAXS) and micro-spot analysis.
figurations were purported to have a limit of detec-
tion (LOD) in the range of 0.5- 2%1,2,3 modern systems Determining Amorphous Content
with specially designed optics can have detection
limits below 0.1%, up to an order of magnitude be- Accurate determination in the 1-10% amor-
low the commonly believed detection limit. These phous content range is now possible.4 PXRD

16
2. partial least squares
(PLS) regression

The PLS method is used by other


techniques as well, such as FT-IR
for pharmaceutical amorphous
determination, and is ideal for
cases where discernible peak(s)
attributed to a phase are not
found in the pattern. “PLS uses the
whole spectrum or a selected re-
gion of the spectrum to develop
calibration models.”5 As shown
in Figure 3, the PLS method pro-
Figure 1a. 0.25% alpha-indomethacin peak measured with BBHD optic; 1b. 0.25% duced a calibration curve with
alpha-indomethacin peak measured with slit optics
a significantly better standard
instrumentation advances now amorphous content range be- deviation (0.035% vs 0.262% for
make it possible to produce high- the peak to background meth-
tween 1 and 10 percent; the re-
er quality data combined with od), higher coefficient of deter-
sulting data set was then analyzed mination (R2) which equates to
modern approaches to the quan-
using two different methods to a better fit of measured to pre-
tification of amorphous content.
quantify the amorphous content: dicted values, and a lower error
Figure 2 shows an example of intercept value (0.024% versus
whereby 10 α-lactose mono- 1. peak to background 0.179%). This shows that PLS can
hydrate standards created with ratio method be a more reliable and accurate
method to determine low amor-
phous content.

Exploring
Co-Crystallization
Parameters and
Polymorphs
The investigation of polymorphs,
chemically identical substanc-
es present in different crystal-
lographic forms, is important in
drug development. APIs often
Figure 2. PXRD patterns for ten α-lactose monohydrate standards with 1-10% occur in a variety of polymor-
amorphous content phic forms, potentially displaying

17
Figure 3. Comparison of confidence limits for two methods of amorphous content determination

different dissolution properties,


and as a result, different bioavail-
ability. To optimize critical prop-
erties of a drug, polymorph anal-
ysis and salt screening may aid in
the selection of a suitable solid
form of a drug. The discovery and
protection of all polymorphs by
patent allows drug inventors to
secure intellectual property and
market position.

PXRD systems equipped with


an X-Y-Z-stage for combinatori-
al screening of samples in a well Figure 4. Cluster analysis of PXRD patterns measured on a 96 well plate
plate can perform high resolu-
tion, high throughput screen- aided by automated cluster analy- and temperature combinations.
ing when using a transmission sis software. Cluster analysis (Figure 4) of the
configuration. PXRD patterns collected from
In this example, cimetidine from
To compare, group and identi- MilliporeSigma was dissolved in each well showed three clus-
fy the crystalline (or amorphous) various solvents and dispensed ters of closely related scans; fur-
phases formed in the wells, the into 96-well plates to recrystal- ther analysis of representative
processing of data is greatly lize under various concentration scans from the clusters could

18
be performed for indexing and stages, allowing the measure- ‘phase’ diagram could be devel-
full pattern fits. Cluster analysis ment of PXRD data at RHs up to oped. Trehalose dihydrate (Th)
greatly aids in the sorting and 95% at room temperature and was seen to be very stable over
analysis of large amounts of data. 75% at 60 degrees C. a wide range of temperatures.
Two polymorphs of trehalose an-
In this experiment, crys-
Stability of Materials hydrate were observed: a stable
talline trehalose dihydrate form Tβ and an unstable form
Subjected to Varying (C12H22O112H2O), an important Tα. An amorphous form of treha-
Heat and Humidity cryoprotectant used in the pro- lose (Ta) was also observed in the
duction of protein-based drugs high temperature – low humidity
Storage and transport of pharma-
that undergo lyophilization, was regime. Important to note is that
ceutical substances can expose
exposed to a range of tempera- once Tβ formed, the phase did
them to heat and/or high relative
ture and relative humidity com- not return to the Th form upon
humidity (RH), which in turn can
binations within a nonambient cooling – a permanent transfor-
induce unexpected phase trans-
stage on a Malvern Panalytical mation occurred.
formations that can affect a prod-
Empyrean X-ray diffraction sys-
uct’s efficacy.
tem. The PXRD patterns were References
Modern PXRD systems can be analyzed to determine which
equipped with the latest tem- phases were present at each 1. http://www.nf-itwg.org/pdfs/ITWG-
perature-controlled humidity condition, and a resultant RH-T INFL-PXRD.pdf, Page 6

2. http://www.xrd.us

3. http://old.vscht.cz/clab/RTG/
dokumenty/panalytical/xrd/X-ray%20
powder%20diffraction%20-%20
a%20powerful%20tool%20in%20
pharmaceutical%20analysis.pdf

4. http://www.
americanpharmaceuticalreview.
com/Featured-Articles/36758-
Approaches-to-Quantification-of-
Amorphous-Content-in-Crystalline-
Drug-Substance-by-Powder-X-ray-
Diffraction/

5. S. Byrn, G. Zografi, X. Chen, Solid State


Figure 5. RH - T phase diagram derived from constant temperature and constant Properties of Pharmaceutical Materials,
relative humidity experiments John Wiley and Sons, 2017, p.337

19
An Interview With...

Andrew L. Theophilus
R&D Manager - Physical Properties
Chemo R&D

During his time in academia, Andrew originally studied biological sciences in Cardiff, specializing in respiratory
physics. In 1991, Andrew joined Glaxo Group Research, formulating and analyzing inhaled medicines. Andrew went
on to specialize in the physical analysis of raw materials and products, expanding into oral and other formulation
types. During his 24 years with Glaxo, Glaxo Wellcome and GlaxoSmithKline, he took on roles of increasing respon-
sibility, ultimately managing a materials science group and representing GSK in various academic collaborations.

Three years ago, Andrew moved to Spain and joined Chemo R&D in Madrid. As R&D Manager - Physical Properties,
he is responsible for defining the physical characteristics of API, excipients and formulated materials, across a broad
portfolio, to ensure consistent product performance.

different medicines. Method development and val-


As someone who has worked on the
idation needs to be efficient. Methods themselves
development of both generic and
need to be rapid, automated if possible, yet still pro-
innovator drug products, have you
ducing high quality data.
noticed any differences between the
physicochemical analysis approaches Generics, it would seem, have the advantage of
used in the generics industry background information on a product, and this is
compared to those used by true to some extent; no doubt there will be patents
innovators, and do you see any and literature relating to the API and product. But as-
sessing this literature throws up problems. Take poly-
opportunities for analytical learnings
morphism, for example. There are sure to be patents
to be shared between the two?
covering numerous polymorphs – I have seen some
One of the main differences I have seen in generic materials with in excess of 20 forms reported. Some
companies is that there are a greater variety of proj- of these are likely to be mixtures of forms, or dupli-
ects and the pace and throughput is much faster cates patented by different companies, and there
compared to innovators. I encounter a large port- are bound to be huge gaps in the available data.
folio of APIs (Active Pharmaceutical Ingredients), Seeing the wood for the trees is tricky. You need to
excipients and final dosage forms across a lot of be a detective and find the clues.

20
For innovators the issues are different. Characterization
You are involved in the development
is much more targeted. More effort can be spent on
of multiple dosage forms. Could you
specific molecules. But it is necessary since you are
comment on the challenges of
starting from scratch with a new material, perhaps
with very little material to work with - just a few mil-
characterizing the physical
ligrams in many cases. It’s likely that each small scale
attributes of such a wide variety
batch will be different to the last, with changes in of products?
particle size, shape, form, water content and so on.
Final dosage forms are often an interesting chal-
The challenge of understanding a material that has lenge. The properties of the API are clearly import-
never existed before is exceptionally rewarding. The ant, but the properties and functionality of the
use of complementary techniques is key. There is no medicine heading for the patient is critical for effi-
one technique that will tell you all you need to know. cacy and safety. The technologies for examining the
X-ray diffraction, thermal analysis, Raman and IR spec- properties of these various dosage forms are limit-
troscopies are the minimum for understanding solid ed in comparison to those for studying API. Various
state, with ssNMR and other techniques to try to fill microscopy-based imaging systems with image
analysis are available, as well as chemical imaging
some of the gaps. Then surface technologies like va-
technologies based on various spectroscopic tech-
por sorption and microcalorimetry if any disorder or
niques. These have improved the situation a great
amorphous material is involved. As more material deal, but they have their limitations, particularly the
becomes available during scale up, particle size and resolution of small particles.
surface area go without saying (but careful develop-
Some of the more tricky formulations relate to
ment of a particle size method is critical, even at this
creams and ointments. Isolating or characterizing
stage). But at every stage the most important tech-
the API, in situ, in complex multicomponent colloidal
nique of all is microscopy. You have to visualize the systems require a variety of different technologies,
material, see it with your own eyes. A picture paints mainly microscopic, and the choice is highly depen-
a thousand words, but microscopy underpins a thou- dent on API concentration and the presence of addi-
sand techniques. tional components. When it is necessary to measure

22
the API particle shape, size and polymorphic form, the performance of a medicine, be it dissolution
it helps to have an arsenal of possible techniques or in vivo testing. If, for example, surface area is
at your disposal. Injectable suspensions present a the better parameter to control tablet dissolution,
similar challenge. Again, analysis in situ would be there is no reason why it shouldn’t be the primary,
ideal, but many techniques are confounded by the or indeed only critical quality attribute. I know that
dispersion media or the glass container. Some anal- regulatory agencies will support this approach if
ysis techniques can be used – Raman is always one
they are provided with a good scientific argument
option as well as optical microscopy, but sometimes
and the data to support it.
isolation from the dispersing medium by drying or
filtration is the only approach. But clearly the con-
sequences of either approach include the possibility Can you comment on the use of any
of damaging or changing the API or crystallizing out emerging technologies which provide
soluble components. additional information on a product’s
physical properties and thus its
Are there any frustrations or areas predicted quality and performance?
for improvement you can identify
with regard to the types of There are some amazing new technologies for the
analytical data required for characterization of APIs and formulated materials.
regulatory submission? There have been great strides forward in imaging
and related technologies such as Raman and X-ray
Certain physical techniques appear, almost by diffraction imaging, surface analysis technologies
default, in regulatory specifications. In the same such as various gas and vapor sorption techniques,
way that identity, assay and impurities will always as well as huge advances in computing power, driv-
feature in a release specification for API, particle ing modelling and prediction technology. But per-
size will normally feature as well, for example. I be- haps we should take time to think about what we
lieve this is driven by an assumption that particle are actually analyzing. It is great that we can ana-
size will always influence processability or tablet lyze samples with higher and higher resolution, and
dissolution, and also by the apparent ease and greater and greater precision, but if the sample we
availability of particle size equipment. In many are analyzing is not representative of the bulk mate-
cases this is right, but particle size is not the only rial we are trying to characterize then these advanc-
characteristic that is important for a product’s es are missing important information. Something
performance, and sometimes it is of very little im- every physical scientist must remember is that the
portance at all. Sometimes particle shape is more materials they work with are heterogeneous, and as
important in processability, and, as theory tells us, such it is important to understand the entire com-
the surface area of an API should be the main driv- position of these materials. For example, the correct
er in dissolution. Correlation with product per- sampling of a 10 kg batch of API, is as important as
formance is the key. It is so important to identify any characterization technology you subsequently
which attributes of a material actually influence apply to it.

23
Comparing Your Material’s
Structure to Literature and
Patent PXRD Patterns
Patents and literature can be sources for digital or
printed diffraction patterns, which may be needed
to compare to material you are producing. A bitmap
converter found in Malvern Panalytical’s Highscore
(Plus) software1 allows digital or printed images to
be digitized into an XML file suitable for diffraction
analysis and comparison. This allows all related pat-
ented polymorph patterns to be added, along with
patterns calculated from single crystal structure
solution data, into one Search/Match database for
comparison purpose to ensure that synthesized APIs
do not infringe patents. In this example (see Figures
1 and 2), images of powder X-ray diffrac­tion (PXRD) Figure 2. Cluster analysis of the patterns found in
patterns in patent US 8217061 B22 were converted US 8217061 B2
into XML files and compared by cluster analysis to
look for similar patterns. The seven patterns formed
two clusters of two similar patterns, and three were
found to be statistically different from the clusters.
Digitization of PXRD data from patents and litera-
ture can be a powerful way to ensure that patent
violation is not occurring and can confirm a new
entity is unique.

References
1. T. Degen, M. Sadki, E. Bron, U. König & G. Nénert, The HighScore
Suite, Powder Diffr. Vol. 29, (2014), Supplement S2, S13 - S18.
Figure 1. Digitized PXRD patterns from
patent US 8217061 B2 2. https://patents.google.com/patent/US8217061B2/en

24
Automated Morphological
Imaging: Deeper Insight into
your Pharmaceutical Formulation
or Product

Fully-automated morphological imaging of a phar- by imaging individual particles and generating a va-
maceutical formulation or product can deliver particle riety of quantitative shape data.
size and shape information in a single measurement,
A concurrent measurement by Raman spectroscopy
and with minimal user intervention. Simultaneous de-
of each particle can provide the complete picture of
termination of chemical composition by Raman spec-
what is present in a multicomponent system, or per-
troscopy delivers the power to unlock complex particle haps what unwanted particle type or contaminant is
characterization problems. present in a product. This powerful combination of
A knowledge and understanding of particle shape as technologies can also be used to speed the defor-
well as particle size is essential in the development, mulation of the most complex pharmaceutical prod-
control and optimization of pharmaceutical formu- ucts, helping to establish in vitro bioequivalence
with the reference product.
lations and final products. Particle characterization
using image analysis-based systems can provide real
insight into the nature of particulate materials, sup-
Why Particle Shape is Important
porting each stage of pharmaceutical development, Even very small differences in particle size or shape
from the selection of raw materials, via formulation can have a significant effect on the properties and
development, right through to QC. From an intel- performance of materials. In pharmaceuticals,
lectual property perspective, this information can for example, bioavailability, flowability, stability,
be used to tighten the specification on a particular blending or tabletting efficiency may all be affect-
product and make it difficult to replicate. This type of ed. Measuring particle size alone may not be sen-
detailed analysis is now easily achievable, thanks to a sitive enough to identify the important but subtle
new generation of automated analyzers, the best of differences between samples. Particles with the
which provide size and morphological information same overall size but very different shapes may

26
be characterized as identical
by some techniques. Applying
image analysis is a highly effec-
tive way of simultaneously mea-
suring particle size and shape.
Furthermore, particle charac-
terization by image analysis is
complementary to other parti-
cle sizing methods such as laser
diffraction. So, not only does it
provide additional information,
it also offers a means of validat-
ing other techniques.

Size and Shape


Definitions
Describing a 3-dimensional par-
ticle can be much more complex
than it first appears. While it can
Figure 1. Key particle dimensions
be convenient to use a single
number, if the particle is not a
that becomes larger or smaller as
perfect sphere there are multi- Sample Preparation
the particle does. Importantly, it
ple ways in which its size could
is both objective and repeatable.
Challenges
be described (Figure 1). Image
analysis captures a 2-dimension- Particle shape is an even more A significant challenge in making
al image of a 3D particle, from complex challenge and requires particle characterization by im-
which it calculates various size the measurement of many pa- age analysis a routine operation
and shape parameters. Principal rameters to build a complete is the need for fast and consis-
among these is circle equivalent picture. The calculation of multi- tent sample preparation. Sample
(CE) diameter, which can be used ple shape parameters, including preparation must be reproduc-
to calculate particle size. The cap- convexity, elongation and so- ible, and samples must be homo-
tured image is converted to a cir- lidity, for every particle, and the geneously distributed, usually on
cle of equivalent area and while generation of distributions for a glass slide or plate which is then
differently shaped particles will each, allows the identification placed on an x-y stage for imag-
influence the CE figure, it has the and quantification of even very ing. Reproducible sample prepa-
virtue of being a single number subtle differences. ration will ensure that results are

27
repeatable, while homogeneity allows the option The first stage of an MDRS measurement is automat-
of analyzing a subsection, knowing that it is repre- ed image analysis of the dispersed sample, to de-
sentative of the whole. This translates into shorter termine the size and an array of shape parameters
measurement times and higher throughput, making for each individual particle, together with its precise
image analysis suitable for routine use. Dry powders position on the measurement slide. This information
pose a particular challenge, requiring strict control is then used to direct the Raman spectroscopic anal-
of dispersion conditions. Reducing sample prepara- ysis, the second stage of the technique.
tion times and eliminating user bias demands a high
At its simplest, Raman analysis is performed on a
level of automation. One highly successful solution
randomly-selected subset of the particles, allowing
is the integration of a sample dispersion unit within
the user to chemically identify a sample’s multiple
the particle characterization system itself.
components, create particle classes based on these
Adding Component-Specific assignments, and determine the relative propor-
tions of each component. A morphological descrip-
Microstructural Insight
tion of each chemically-identified component can
Identification of the different components with- then be generated in the form of size and shape
in a formulation or product completes the particle distributions, using the data from the automated
characterization picture. Morphologically-Directed image analysis.
Raman Spectroscopy, or MDRS®, combines the tech- If particular components in the blend are of interest,
niques of automated particle imaging and Raman the measurement can be refined by selectively tar-
microspectroscopy in an integrated platform, to en- geting those particles with a distinct morphology
able rapid, automated chemical and morphological for chemical analysis; for example, particles within a
characterization of the individual components in certain size range, or with shape parameters above a
a multi-component sample. Multiple applications specific value. By focusing the Raman measurement
benefit from this, including: only on the components of interest, more of these
• Establishing the particle size of the active particles can be characterized in a given timeframe.
pharmaceutical ingredient (API) within a
formulation Accelerating Bioequivalence
• Understanding the impact of process steps or
Assessments
changes on the API or excipients The application of MDRS in support of in vitro bio-
• Pinpointing the origin and identity of any equivalence studies has recently been shown in the
contaminants approval of an abbreviated new drug application
(ANDA) for a locally-acting mometasone furoate na-
Comparing the particle size and shape of the API in
sal spray.1
reference listed drug (RLD) or generic (test) formula-
tions – useful during deformulation and when inves- MDRS enables determination of the size and shape
tigating in vitro bioequivalence of specific components within a multi-component

28
Figure 2. Use of MDRS in establishing particle identity, componentspecific particle size distribution, and comparability
between reference and test formulations.

blend. For the purposes of the ANDA, this capabil- measurement and data analysis capabilities, is al-
ity was used to compare the form and particle size lowing the more widespread use of this technique
of the API in the reference and test nasal sprays and research, development, and also quality assurance.
confirm their close similarity as seen in Figure 2. Automated sample preparation, integrated as part
The resulting data from a Malvern Panalytical of the measurement process, has proven to be one
Morphologi system were accepted in lieu of a clinical of the major keys to even higher throughput with
bioequivalence (BE) endpoint study, setting a prece- excellent reproducibility, especially for dry powders.
dent for the inclusion of in vitro studies of bioequiv- The integration of complementary technologies,
alence in future ANDAs – this could help reduce the such as Raman spectroscopy, provides the compo-
cost and complexity of tests associated with a sub- nent-specific microstructural insight which is crucial
mission and accelerate new products to market. for a deeper understanding of both pharmaceutical
products and processes.
Conclusion
References
Knowledge and understanding of particle shape as
well as size is now essential for many applications. 1. FDA/CDER SBIA Chronicles; FDA embraces emerging technology
The advent of automated image analysis-based for bioequivalence evaluation of locally-acting nasal sprays:
particle characterization systems, with powerful https://www.fda.gov/media/97705/download

29
An Interview with
Paul Kippax
Pharmaceutical Sector Director at Malvern Panalytical

me to move on to managing the development of


Can you tell us a little about your
the company’s Mastersizer laser diffraction parti-
journey with Malvern Panalytical?
cle sizing system and, finally, to leading the prod-
I came to Malvern Instruments, as Malvern uct management group associated with Malvern
Panalytical was then known, almost 23 years ago, Panalytical’s complete portfolio of morphological
following the completion of my Ph.D. in physical characterization solutions.
chemistry and colloid science. My first role was We’ve long known that understanding our custom-
as an applications engineer, but I moved into our ers’ challenges is key to helping us understand where
Product Management department in 2002, and to focus and innovate. Over the past couple of years,
stayed there for 16 years!
we’ve made great strides in this area and grown into
I’d always been interested in the application of par- a truly customer-centric organization by creating
ticle characterization technologies, but it was our dedicated teams focused on developing deep un-
Spraytec spray particle and droplet sizing system, derstanding of the industries we serve. As a result of
and an understanding of how it was being used this, I jumped at the chance to move into my current
in nasal spray and inhaler product development, role as Pharmaceutical Sector Director, which, from
which first introduced me to the pharmaceutical in- the beginning of this year, has enabled me to con-
dustry. I recall attending the UK’s Drug Delivery to centrate on pharmaceutical industry applications.
the Lungs conference back in 1999, where I heard
how our customers were struggling to find a way to How does Malvern Panalytical ensure
control drug delivery from aerosol-based devices. it stays close to its customers and
Realizing that Spraytec could help, I started work- understands their needs?
ing on collaborative projects to see if we could gen-
erate the relevant data to understand drug prod- We’re constantly improving the way in which we work
uct performance. The experience I gained enabled with our customers and, through this, developing

30
our understanding of how we can support them in development of our Morphologi 4 range of an-
being more effective or generating additional value alytical imaging solutions. Understanding the
from their own projects. market’s desire to establish the bioequivalence of
formulations in vitro, and also hearing direct feed-
We’ve always had an active account management
back from the US FDA, helped us to recognize the
program, where we partner with key customers.
relevance of the Morphologi 4-ID, which provides
Here, we aim to become a trusted member of a cus-
tomer’s team, maintaining open lines of communica- component-specific chemical identification. We
tion and support. Often this has been based on the then knew how to build on the capabilities of this
supply of a single instrument or solution within their technology so it could be applied more effectively
organization. However, we’ve always understood within our customers’ development projects.
that our technologies are not stand-alone but form For another of our solutions launched last year, the
part of our customers’ workflows alongside other
Zetasizer Ultra, we received significant feedback on
analytical methods. We have therefore always tried
our proposed analytics workflow from customers
to give good advice on method selection in line with
very early on in development. Some of our custom-
our customers’ product and process development
ers then helped us beta-test the product, which was
and control requirements, even when this involves
a huge help with the prioritization of its software
specifying solutions we do not provide! This ensures
development, shaping the solution into the success
that when our solutions are applied, this is done in a
story it is today.
way which enables the identification of relevant crit-
ical material properties, enabling a QbD approach to
product development.
How does Malvern Panalytical decide
which technologies to develop next?
We are heavily engaged with our academic net-
work and often present applications work at sci- We’ve been engaged in providing analytics to the
entific conferences. We do this not to just promote pharmaceutical industry for many years now, so
– we aim to provoke a response from the audience we keep a close eye on industry trends. During the
and gain more insight into how our solutions are 1990s, the use of Malvern Panalytical technologies
currently applied or could be developed in the fu- within the pharmaceutical industry grew dramati-
ture. Here, we’re interested in the realistic, relevant cally, as the importance of microstructure in defin-
and valuable application of our technologies – and ing product and process performance increased.
where they can be used synergistically. That growth gave us the foundation to provide our
As an organization, we involve our customers and userbase with a commitment that we would ensure
key opinion leaders is in the development of our consistent access to the capability to measure crit-
solutions – testing prototypes of software and ical material attributes such as particle size – this
measurement workflows and helping us under- translates into a promise to continually support our
stand what’s most important to them. Recently, customers, which feeds into how we maintain our
we’ve worked directly with our customers in the solution portfolios.

31
When we start to consider what drives our direc- in vitro bioequivalence studies. We’re also work-
tion, there are several elements at play. ing with our customers to understand how micro-
Firstly, we’re seeing an increase in interest in data in- structural understanding can be applied in innova-
tegrity. This has been prompted by regulatory guid- tor product development workflows to speed the
ance such as the US FDA’s 21 CFR Part 11 rule, relat- identification of candidate formulations and en-
ing to electronic records and signatures. However, able new products to be brought to market more
data integrity goes deeper than simply considering quickly.
how data is stored; it also challenges whether the
measurement data itself is relevant to controlling Finally, we value working with our customers to
product quality and whether it has been generat- guide us on appropriate routes forward. In fact, this
ed in a product-relevant way. So, although applica- works both ways! We’ve learned how our analytical
tion of data integrity principles is often considered solutions fit alongside other techniques to guide
a QC issue, it actually extends all the way back to new product development. We know that the solu-
decisions made and analytics applied during the tions we provide are being used to help eliminate
development process. Our customers’ focus is to drug candidates or candidate formulations that are
consider what is required for the patient and the unlikely to be viable at a much earlier point in de-
treatment of a given disease. Malvern Panalytical
velopment than was possible in the past, optimiz-
can help with the selection of relevant physico-
ing and speeding the drug development pipeline.
chemical measurement parameters and methods
to ensure that realistic and reliable data are gener- Alongside this, we recognize the pressure on our
ated to guide good development decisions, which customers to rapidly assimilate multiple analytical
in turn ensure that the product is safe and effective. datasets in order to inform their development de-
We’re also very aware of global requirements to cisions. With the desire to compress product devel-
drive down the cost and increase the availability of opment times, and the requirement for scientists
both existing and new medicines. Important within to support multiple concurrent projects, there is
this is encouragement of the development of more often not enough time for our customers to devel-
generic products, particularly for complex drug op and retain deep knowledge regarding the appli-
products and formulations. Regulators are consid- cation of specific techniques. We therefore partner
ering the application of in vitro approaches to as-
directly with development teams in order to aid
sessing bioequivalence as a surrogate for clinical
realistic method selection. Here, the recent addi-
endpoint studies, in order to aid generic product
tion of the contract research organization Concept
development. These approaches consider the im-
portance of the microstructure of the dosage form Life Sciences to the Malvern Panalytical platform is
in defining the effectiveness and reproducibility helping us on many levels: from understanding the
of drug delivery. Malvern Panalytical’s solutions industry and its requirements to implementing a
provide relevant data to support and direct these given technology in our customers’ workflows.

32

You might also like