You are on page 1of 8

reviews research focus DDT Vol. 8, No.

19 October 2003

Solid-state analysis of the active


pharmaceutical ingredient in drug
products
Ann W. Newman and Stephen R. Byrn

The solid form of a drug substance is important when developing a new Considerations for solid-state during
chemical entity. The crystalline form used in development is significant development
The delivery system to be targeted is an im-
based on possible manufacturability, solubility, bioavailability and stability
portant consideration when considering solid-
differences between the solid forms. Regulatory issues require that the state issues during development. A variety of
form present in a solid dosage form or liquids containing undissolved drug dosage forms are available for pharmaceutical
substance be identified. Drug product samples can be analyzed by a products (for examples see Box 1). The key
solid-state parameters for the drug substance
variety of techniques to determine the crystal form present or changes
in a tablet formulation will be significantly
that occur during the manufacture of a drug product. The form present different than those for an oral suspension or
will affect development, regulatory and intellectual property issues. inhalation product.
Regulatory issues for solid dosage forms or
▼ The solid form of a drug substance, or ac- liquids containing undissolved drug substance
Ann W. Newman
SSCI, West Lafayette tive pharmaceutical ingredient (API), is im- are outlined in an International Conference
IN 47906, USA portant when developing a new chemical en- on Harmonisation (ICH; http:// www.ich.org)
e-mail: anewman@ssci-inc.com tity (NCE). Crystalline organic solids are made decision tree, as shown in Figure 2 [2,3]. The
Stephen R. Byrn
Purdue University
up of molecules that are packed or ordered in polymorph or the crystal form used to
West Lafayette a specific arrangement. These molecules are produce the dosage form must be known, as
IN 47907, USA held together by relatively weak forces, such should how crystal form changes might affect
e-mail: stephen.byrn@gte.net
as hydrogen bonding and van der Waals in- the performance or stability of the drug prod-
teractions. The arrangement of the molecules uct. These are important questions that must
is defined by a unit cell, which is the smallest be answered during the drug development
repeating unit of a crystal. These different process.
arrangements determine the crystal form of The information obtained from drug prod-
the material and can include solvates, hy- uct analysis is important in many areas. When
drates and unsolvated materials. The term designing formulations, it is imperative to
polymorph describes multiple crystal forms know which crystal form of a drug is present
with the same molecular formula; for exam- at the various stages of a process and at the
ple, two unsolvated forms are considered end of the process. A hygroscopic drug will
polymorphs, just as two monohydrate forms likely form a hydrate during a wet granu-
are considered polymorphs but an unsolvated lation, but that hydrate could also convert or
and a hydrated form are not considered poly- partially convert to another form upon dry-
morphs. A noncrystalline solid is called amor- ing. A direct compression formulation might
phous, where the molecules are not ordered be feasible but only under certain relative
in a specific arrangement (Figure 1). The crys- humidity conditions. It is important to deter-
tal form used in development is important mine if there are phase transformations
based on possible manufacturability, solubil- occurring during processing, as well as what
ity, bioavailability and stability differences crystal form is present in the final drug
between the different solid forms [1]. product [4–7].

898 www.drugdiscoverytoday.com 1359-6446/03/$ – see front matter ©2003 Elsevier Science Ltd. All rights reserved. PII: S1359-6446(03)02832-0
DDT Vol. 8, No. 19 October 2003 research focus reviews

Another consideration is possible


crystal form transformations over time
or during solid-state (also known as
physical) stability testing. This is not
the same as chemical stability, which
deals with degradation; however, the
same samples can be used and analyzed
by different methods to determine
physical and chemical stability. In many Order (crystalline) Intermediate order Disorder (amorphous)
solid-state systems, two polymorphic
Drug Discovery Today
forms can exist that have different
thermodynamic stabilities. The more Figure 1. An illustration of the concept of molecular order in solids.
stable form is usually chosen for devel-
opment but a solubility advantage
could be obtained with the less stable form. In such
Box 1. Examples of pharmaceutical dosage
systems, the less stable form can transform to the more
forms
stable form especially under the influence of heat,
mechanical stress, moisture or seeds. Establishing the solid- Tablets Patches
state stability (no crystal form change) under various Emulsions Capsules
conditions will help determine the shelf life of the drug Depot Inhalers
product or force the development of the more stable form Solutions Implants
Suppositories Suspensions
if the metastable form does not exhibit sufficient stability.
Intravenous (IV) Soft gel capsules
The importance of solid analysis of API in drug product
development will be discussed in the following sections,
including common techniques used to analyze solid exhibit peaks indicative of reflections from specific atomic
dosage forms, examples of drug product analysis and process- planes; these patterns are representative of the structure
induced transformations during formulation. but do not give positional information about the atoms in
the molecule. One peak will be exhibited for all repeating
Techniques planes with the same spacing. By contrast, an amorphous
Several analytical techniques are commonly used in form- sample will exhibit a broad hump in the pattern called an
selection studies. Various publications [8–10] and books amorphous halo. Representative XRPD patterns for lactose
[1,11] describe physical characterization of solid-state are shown in Figure 3.
pharmaceuticals. A brief description of common methods Qualitative analysis of powder patterns can be used to
will be presented here. determine if multiple samples are the same crystal form or
if multiple crystal forms have been developed. Mixtures of
X-ray powder diffraction samples can also be evaluated. When mixtures are ob-
X-ray diffraction techniques used for characterizing phar- tained, XRPD can also be used in a quantitative mode to
maceutical solids include the analysis of single crystals and calculate the amount of each phase present.
powders. The electrons surrounding the atoms diffract
X-rays in a manner described by the Bragg equation [11]. Vibrational spectroscopy
X-rays will be diffracted at an angle (defined as θ), and this Common techniques used to characterize drugs and excip-
knowledge, together with knowledge of the X-ray wave- ients are infrared (IR) and Raman spectroscopy, which are
length, can be used to calculate the spacing between the sensitive to the structure, conformation and environment
planes. of organic compounds. Because of this sensitivity, they are
X-ray powder diffraction (XRPD) is the analysis of a useful characterization tools for pharmaceutical crystal
powder sample with a typical output being a plot of inten- forms. Qualitative as well as quantitative analysis can be
sity versus the diffraction angle (2θ). The value 2θ is used performed with both techniques.
based on the configuration of the instrument. Such a plot IR spectroscopy is based on the conversion of IR radi-
can be considered a fingerprint of the crystal structure and ation into molecular vibrations. For a vibration to be IR
is useful for determining the crystallographic similarity of active, it must involve a changing molecular dipole (asym-
samples by pattern comparison. A crystalline material will metric mode). For example, vibration of a dipolar carbonyl

www.drugdiscoverytoday.com 899
reviews research focus DDT Vol. 8, No. 19 October 2003

crystal form determination because


minimal sample manipulation is re-
Monitor polymorph change on Can
No No need to set acceptance quired. Mulls can also be used for form
manufacture or stability of polymorph ratio criteria fpr polymorph
drug product change? content in drug product identification, but peaks that result
from the suspension medium could
interfere with the peaks of interest.
Yes Raman spectroscopy is based on the
inelastic scattering of laser radiation
Does
drug product
with loss of vibrational energy by a
performance No need to set acceptance sample. A vibrational mode is Raman-
testing provide Yes
criteria for polymorph active when there is a change in the
adequate control content in drug product
(e.g. polarizability during the vibration.
dissolution)?
Symmetric modes tend to be Raman
active; for example, vibrations about
No
bonds between the same atom, such as
Establish acceptance criteria
in alkynes, can be observed by this
that are consistent with technique.
safety and efficacy
Small amounts of samples can be
Drug Discovery Today
analyzed by Raman spectroscopy and
Figure 2. Regulatory decision tree for solid dosage forms or liquids containing a variety of sample holders are avail-
undissolved drug substances (adapted from Ref. [3]). able, ranging from stainless steel hold-
ers to glass NMR tubes. The samples
are analyzed neat, eliminating the
need for sample preparation proce-
dures that might induce solid form
changes. A laser is used, therefore, only
a small portion of the sample is in the
beam during analysis.
Both IR and Raman microscopes are
available for imaging drug product
samples [12–14]. Several methods of
analysis, such as spot mode, line scan
or area mapping, can be used to look at
the distribution of a crystal form in a
dosage form, such as a tablet, or to find
minor levels of a form. Recently, NIR
microscopy has been introduced as a
powerful new method for drug product
analysis by imaging [13].

Drug Discovery Today


NMR spectroscopy
Figure 3. XRPD patterns of crystalline anhydrous lactose (top) crystalline lactose NMR spectroscopy probes atomic
monohydrate (middle) and amorphous lactose (bottom). environments based on the different
resonance frequencies exhibited by
nuclei in a strong magnetic field.
group is detectable by IR spectroscopy. Whereas IR has Many different nuclei are observable by NMR but those of
been traditionally used as an aid in structure elucidation, hydrogen and carbon atoms are the most frequently stud-
vibrational changes also serve as probes of intermolecular ied. NMR spectroscopy of solutions is commonly used for
interactions in solid materials. structure elucidation. However, solid-state NMR measure-
Sampling techniques for IR include pellets, mulls and ments are extremely useful for characterizing the crystal
diffuse reflectance. Diffuse reflectance is the best choice for forms of pharmaceutical solids.

900 www.drugdiscoverytoday.com
DDT Vol. 8, No. 19 October 2003 research focus reviews

Table 1. Summary of quantitative methods


Quantitative method Scope of assay Result
Limit test A limit of detection for a technique is Result is specified as ’2% Form B present’
determined (such as 2%)
Specification assay An assay limit is determined for a Result is specified as ’95% Form A present’
technique (such as 95%)
Full quantitative method A minimum quantifiable limit (MQL) (such 'X% of Form B' if within the quantitation range of
as 2%) and linear range (such as 2–25%) 2–25% or ‘2% Form B' if below MQL or '25% Form
is determined B' if above linear range

Nuclei that are typically analyzed with this technique in- are outlined in Table 1. Various levels of validation are re-
clude those of 13C, 31P, 15N, 25Mg and 23Na. Different crystal quired for the different methods. Methods become more
structures of a compound can result in perturbation of the complex as the number of contaminating forms increases.
chemical environment of each nucleus, resulting in a
unique spectrum for each form. Once resonances have been Analysis of the drug product
assigned to specific atoms of the molecule, information on There have been several reports on the analysis of solid
the nature of the polymorphic variations can be obtained. dosage forms [16–28]. A variety of techniques, such as
This can be useful early in drug development when the sin- XRPD [16–23], NMR [24–27], Raman [28–29] and IR [30]
gle crystal structure might not be available. Long data acqui- have been used to identify the crystal form in a wide
sition times are common with solid-state NMR, so it is often selection of dosage forms, including tablets, capsules, oint-
not considered for routine analysis of samples. However, it is ments, suppositories and microspheres. A small selection
usually a sensitive technique and sample preparation is min- of the available literature will be discussed here.
imal. NMR spectroscopy can be used either qualitatively or
quantitatively, and can provide structural data, such as the X-ray powder diffraction
identity of solvents bound in a crystal. XRPD is a common choice for pharmaceutical dosage form
analysis because it is a direct measure of the crystal form of
Qualitative versus quantitative analysis a material. Overlap of the crystalline API peaks with crys-
Different levels of analysis are used at different stages of talline excipients is a common problem when analyzing
the development process. At the early formulation and drug product samples. However, when the excipients are
process evaluation stage, early Good Manufacturing amorphous, or there is a high loading of the drug in the
Practices (GMP) method development and validation ap- formulation, XRPD can be an effective technique in identi-
proaches are used. Qualitative or visual determinations can fying crystalline API. Qualitative or quantitative [16,17]
be made at this stage instead of full quantitative analyses. determinations can be performed using this technique.
These measurements are used to guide the development of Tablets and capsules are common dosage forms to ana-
the drug and might not be applicable to the final chosen lyze. An example is the analysis of marketed ampicillin cap-
form. During Phase II, stronger GMP method development sules [17] where the anhydrous or trihydrate forms were
and validation are needed. These methods are developed readily identified by XRPD at a drug loading of ~83% w/w
in preparation for end-of-Phase II meetings where ques- ampicillin. Dosage forms with multiple active ingredients
tions about method accuracy and suitability could arise. can also be effectively analyzed, as demonstrated using
Quantitative or limit test methods are usually in place to marketed Excedrin® tablets. Identification of acetamino-
aid early process validation. By the product launch, final- phen and aspirin was readily performed; however, further
ized methods and full validation of methods and the data analysis using pattern subtraction was required to
process are required. ICH Q2B Guidelines are available for identify caffeine. Quantitation of clinical trial samples of
quantitative methods [15]. dOTC, an antiviral drug in the nucleoside analog class of
There are several quantitative approaches for solid-state drugs, which was previously in development, was per-
testing. For all assays, specificity needs to be determined for formed at SSCI (http://www.ssci-inc.com) using XRPD. The
the technique being used for method development. As an ex- method was developed and validated from 3–30% for the
ample, Form A is the form chosen for development and Form metastable Form A in the desired stable Form B formulation
B is a contaminating form. Possible choices for quantitation and amounts up to 50% were estimated with a second

www.drugdiscoverytoday.com 901
reviews research focus DDT Vol. 8, No. 19 October 2003

calibration curve. Large amounts of Form A (up to 38%) another. The dose of 5mg in a 100 mg tablet demonstrates
were measured for the samples. It was found that over a six the levels that are detectable using this technique.
month time period, the amount of Form A decreased from
~38% to ~30%. This was important information for the Raman and IR spectroscopy
clinical scientists when reviewing the bioavailability data. Raman spectroscopy has also been used to investigate API
The lack of crystalline API peaks when a dosage form is present in tablets [28,29]. The benefits of Raman spectroscopy
analyzed could indicate that the material is amorphous or include minimal, if any, sample preparation, as well as less
that the loading is too low to detect using the parameters overlap with common excipients when compared to IR spec-
chosen. Synchrotron radiation can be used to increase sen- troscopy. The shortcomings include possible fluorescence, a
sitivity and measure much lower levels of different forms small sampling area due to the size of the laser beam used for
in the drug product, as evidenced by data collected on excitation, and possible sample heating during data collection.
ranitidine HCl and Children’s Advil© [18]. Different data Raman spectroscopy has been used to try to identify
analysis methods, such as spectral subtraction [17], counterfeit drug products and was found to be more effec-
Rietveld analysis [18,19], whole pattern matching software tive than IR for the cases presented [29]. In another study,
[20,21] and partial least-square (PLS) methods [22], can tablets of various marketed products were analyzed to
also be used to obtain lower detection levels or quantitate specifically look at the crystal form present in various sam-
materials with significant overlap. ples [30]. In the case of a 2.5 mg prednisolone tablet
Analysis of other dosage forms using XRPD has also been (~1.4% of drug), the peaks for Form II prednisolone were
reported. The amount of crystalline tolnafate was deter- clearly evident. Warfarin sodium exists as an amorphous
mined in ethylcellulose microspheres [23]. Acetaminophen solid or crystalline clathrate of the drug and isopropyl al-
suppositories containing hydrogenated vegetable oil, poly- cohol. A 2.1% loading in a 2.5 mg tablet was analyzed and
oxyethylene stearate, glycol monostearate and preserva- found to contain the crystalline clathrate and not the
tives were examined [17]. Interference with the excipients amorphous solid. In the case of ranitidine HCl, tablets
was evident between ~15°2θ and 25°2θ, however, aceta- from different manufacturers were found to contain different
minophen peaks were clearly present. Pattern subtraction crystal forms. Artonil tablets contained Form I ranitidine
was again used to unequivocally identify the aceta- HCl and Zantac® tablets contained Form II. Raman was
minophen. In the case of a zinc oxide ointment, the also used to distinguish the anhydrous and monohydrate
crystalline inorganic peaks attributed to zinc oxide were forms of theophylline in tablets stored at 75% relative hu-
observed above 30°2θ, eliminating overlap with the amor- midity (RH) for up to 12 days. It was found that the anhydrous
phous halo observed below 20°2θ [17]. form readily converts to the monohydrate form over time.
A near infrared (NIR) method was developed and vali-
Solid-state NMR spectroscopy dated to determine the amount of crystalline miokamycin
Solid-state NMR spectroscopy has become a popular in the presence of the desired amorphous material in mar-
method of analyzing dosage forms [24–26]. The technique keted tablets. A PLS method was developed to quantitate
offers little interference from many excipients but the data crystalline material in the range of 1–9%.
acquisition can be complicated and lengthy. Quantitation
can also be performed with NMR spectroscopy [25,27]. A Chemical imaging
number of parameters need to be determined for each sys- Chemical imaging is a powerful method for the analysis of
tem for the signal intensities to be representative of the drug products. Most Raman studies involve microtoming or
number of nuclei being measured. using some other technique to obtain a smooth surface. It
Several marketed tablets and capsules were analyzed by is possible to image an entire tablet by successively micro-
NMR to determine the feasibility of using this technique for toming and collecting data in that plane. An NIR imaging
solid dosage form analysis [24]. Samples of enalapril maleate system has been developed that is faster than the Raman
(Vasotec®) and ibuprofen (Nuprin®, Haltran®) tablets readily procedure and requires less sample handling [13]. NIR
exhibited peaks due to the drug substance, showing that imaging has also been used to analyze blend homogeneity
NMR can easily be used to differentiate between drug prod- and particle size changes in tablets. Chemical imaging pro-
uct and placebo samples. Analysis of lovastatin tablets vides another powerful method for drug product analysis.
(Mevacor®) resulted in interference from the excipients,
therefore a determination of form using this technique was Changes during processing
not possible. Two prednisolone products analyzed by NMR The presence of different physical forms in various dosage
were found to contain Form I in one case and Form II in formulations could be a result of the initial API that was

902 www.drugdiscoverytoday.com
DDT Vol. 8, No. 19 October 2003 research focus reviews

used to manufacture the drug product


or to changes that occurred during pro- Wetting Drying Processing/storage
(moisture/stress/time)
cessing. Form changes can occur at vari-
ous stages of the formulation process,
Meta-
including wet granulation, drying (in- stable
cluding lyophilization), compression, form
Stable
handling and storage (including stabil-
form
Stable
ity testing) [4,6,7]. Examples of possible
form
changes that could occur during Stable
Hydrate
form
the formulation process are shown Hydrate
Isomorphic
schematically in Figure 4 [6]. desolvate
A change in crystal form to a more
stable, less soluble form can have dire Meta-
Amorphous
consequences in the formulation. A re- solid stable
form
cent example is the semisolid capsule
formulation of ritonavir [31]. During Drug Discovery Today
development, only one form of riton-
Figure 4. Possible form changes during a wet granulation process. (Reproduced, with
avir (Form I) was known even though
permission, from Ref. [6]).
attempts were made to find other
forms. The semisolid formulation was
nearly saturated with respect to Form I ritonavir and ended and 75% RH conditions, the material converted back to
up being supersaturated with respect to Form II ritonavir, crystalline form. It was found that dissolution of capsules
resulting in crystallization of the more stable Form II in the containing crystalline material in the formulation was the
marketed capsules. This change in form resulted in a mar- same as capsules containing the amorphous formulation.
ket crisis until a new formulation with the much less solu- Based on XRPD data, it appears that contact with water at
ble Form II was produced. It should be noted that Form II the dissolving surface of the crystalline form results in
was not observed during development of the API or formu- amorphous or liquid crystalline material.
lation or for two years after the launch of the product. Several studies with theophylline have shown a conver-
Common steps in the formulation process where form sion of anhydrous Forms I and II to the monohydrate in
changes can occur will be discussed here; however, it the presence of water [34,35]. A significant decrease in dis-
should be noted that form changes can occur at almost any solution is observed when the hydrate material is present.
point in the process. Other studies have shown that wet granulation and fluid-
bed drying will result in a third unsolvated polymorph of
Wet granulation and drying: the influence of moisture theophylline with different dissolution characteristics
and heat [6,35]. Phase transformations to metastable forms can also
Wet granulation is a frequent way of creating new forms, occur. In the case of fosinopril sodium, rapid drying from a
especially if hydrated forms are already known for the drug granulation fluid containing alcohol showed conversion
substance. Testing materials in the presence of water or to the metastable unsolvated Form B instead of the stable
aqueous solvents can help determine the feasibility of unsolvated Form A. The amount of conversion depended
using wet granulation. In the case of cimetidine, anhy- on the drying conditions used [6,36]. It is important to an-
drous Form A was found to convert to hydrated Form B in alyze samples at various stages of a wet granulation and
water over a period of up to 12 months [32]. A mixture of drying process to investigate any form changes occurring
anhydrous Forms A and D were found to convert to hydrate under these conditions.
Form C in water but to Form D in a 50:50 ethanol:water
solution. It was postulated that in a high shear mixer other Compression: the influence of mechanical force
mechanisms would likely increase the solid form transfor- Tablets are one of the most common dosage forms used in
mations. In the case of an HMG-CoA reductase inhibitor pharmaceuticals and form changes upon compression is
(SQ33600), dry blending the formulation resulted in no an active area of study [32,37–39]. Several investigations
change in crystal form; however, wet granulation trans- have been performed on chlorpropamide [40–42]. It is pos-
formed the crystalline material into amorphous [33]. tulated that chlorpropamide Form A and C will partially
When the amorphous formulation was exposed to 33% convert to a highly unstable amorphous material upon

www.drugdiscoverytoday.com 903
reviews research focus DDT Vol. 8, No. 19 October 2003

compression and a form change occurs from the amor- Aerosol formulations are also at risk. A nedocrimal
phous material. It was observed that the stable Form A will sodium aerosol formulation was stored at various RH condi-
convert to the metastable Form C upon compression. tions [46]. From 12–76% RH, the trihydrate form remained
Compressing the material at elevated temperatures will unchanged and there was no change in the in vitro aerosol
produce more of Form C. Compression of Form C at ambi- performance of the drug. When stored at 86% RH, the trihy-
ent temperatures results in the formation of Form A; drate converted to the heptahemihydrate and a significant
however, at elevated temperatures, Form C is maintained. decrease in the deaggregation performance was observed.
Compression pressures will also affect the conversion to
another form [38]. In the case of sulfabenzamide stable Process analytical technology (PAT)
Form A, higher compression pressures resulted in greater Industry and regulatory agencies are seeking ways to im-
formation of the metastable Form B material. Maprotiline prove manufacturing efficiency by increasing throughput,
HCl exists as a stable Form II as well as metastable Forms I reducing costs and improving product quality. New meas-
and III. Upon compression, Form III will convert to stable urement technologies have been developed to acquire, in
Form II, however, Form I will remain as Form I under the real time, information rich data on process performance
same conditions. Cimetadine Forms A, B, C and D will not using on-line or in-line sensors. Process Analytical Tech-
change upon compression but the amorphous material nology (PAT) is a new initiative to enable effective in-
will crystallize under similar pressure conditions [32]. process control during routine manufacture [47,48]. The
goals are to reduce waste, shorten manufacturing time, de-
Storage liver significant improvements in quality consistency and
Storage conditions can have a crucial role in form stability reduce costs. The data collected real-time during the manu-
[5]. The effects of moisture and temperature can readily facturing steps will decrease post-production analysis
cause transformation between hydrated and anhydrous through an improved understanding of the manufacturing
species but temperature alone can also induce solid trans- processes. PAT methods are sensitive to hydration, such as
formations between unsolvated polymorphic forms. determining the drying endpoint for a granulation in a
Recent studies at Purdue University (http://www.purdue. fluid dryer with NIR, and sensitive to solid form, such as de-
edu), as well as published reports [31], have shown that termining a form change during a wet granulation process.
seeding also accelerates transformations. Suspensions are
also at risk for form transformations due to possible disso- Chemical stability
lution and recrystallization phenomenon, especially at Chemical stability in dosage forms is also a concern
elevated temperature conditions. and initial excipient compatibility studies are usually per-
For theophylline, it was found that the tabletting pres- formed during development of the dosage form [49]. It
sure significantly affected the hydration kinetics in the should be recognized that different forms can have differ-
tablet [43]. Hydration was found to significantly decrease ent chemical stabilities. It is often the case that amorphous
with an increase in tabletting pressure. The data suggested materials will not only be less physically stable (prone to
that the hydration of the tablets does not depend on the crystallization), but also less chemically stable. The glass
hydration mechanism of the bulk powder but on the struc- transition temperature and how it changes with moisture
ture of the dosage form. In another study, the conversion content, related to mobility, is an important factor for the
to the monohydrate was manifested as distinct needles on stability and formulation of amorphous materials [50].
the surface of the tablets after storage [44]. A significant Formulation can accelerate chemical degradation as a re-
decrease in the rate of theophylline release was observed in sult of: (1) interaction with excipients, (2) processing
tablets containing the monohydrate needles. The exam- effects and (3) induction by excipients (but not involving
ples given here deal with crystalline and hydrated species chemical reactions with the excipients) [51]. Various
and it should be noted that amorphous materials might reactions, such as oxidation, cyclization and hydrolysis,
also readily convert upon storage. Attempts to prepare a are common degradation pathways and excipient interac-
fast-dissolving form of nifedipine involved using 2-hydrox- tions, such as acid and Maillard reactions, also need to be
ypropyl-β-cyclodextrin to stabilize the amorphous material considered during formulation development.
[45]. When the mixture was heated to low temperatures,
crystalline nifedipine Form A was produced, however at Conclusions
high temperatures, Form B was produced. Significant con- Solid-state analysis of API in drug products covers a wide
trol of storage conditions is sometimes needed to maintain variety of topics, ranging from the form present in the final
the desired solid state form. dosage form to possible changes that can occur upon

904 www.drugdiscoverytoday.com
DDT Vol. 8, No. 19 October 2003 research focus reviews

processing. It is important to note that form changes can 25 Tishmack, P.A. et al. (2003) Solid-state nuclear magnetic resonance
occur at almost any stage during formulation depending on spectroscopy-pharmaceutical applications. J. Pharm. Sci. 92, 441–474
26 Reutzel-Edens, S.M. and Bush, J.K. (2002) Solid-state NMR spectroscopy
your solid-state system. Several analytical techniques are of small molecules: from NMR crystallography to the characterization
available for determining the crystal forms present in the of solid oral dosage forms. Am. Pharm. Rev 5, 112–115
drug product. These techniques can be used for qualitative 27 Bugay, D.E. (1993) Solid-state nuclear magnetic resonance spectroscopy-
theory and pharmaceutical applications. Pharm. Res. 10, 317–327
or quantitative testing. It is important from a regulatory, as 28 Taylor, L.S. and Langkilde, F.W. (2000) Evaluation of solid-state forms
well as an intellectual property, perspective to know the present in tablets by Raman spectroscopy. J. Pharm. Sci. 89, 1342–1353
crystal form(s) present in the marketed drug product. 29 Enculescu, A.L. and Steiginga, J.R. (2002) Raman spectroscopy-
a powerful tool for non-routine analysis of pharmaceuticals. Am.
References Pharm. Rev. 5, 81–88
1 Byrn, S.R. et al. (1999) Solid-State Chemistry of Drugs, SSCI 30 Blanco, M. and Villar, A. (2003) Development and validation of a
2 Byrn, S. et al. (1995) Pharmaceutical solids: a strategic approach to method form the polymorphic analysis of pharmaceutical preparations
regulatory considerations. Pharm. Res. 12, 945–954 using near infrared spectroscopy. J. Pharm. Sci. 92, 823–830
3 Q6A Specifications, Test procedures and acceptance criteria for new 31 Chemburkar, S.R. et al. (2000) Dealing with the impact of ritanovir
drug substances and new drug products: chemical substances. (1997) polymorphs on the late stages of bulk drug process development.
Fed. Reg. 62, 62889–62910 Org. Proc. Res. Dev. 4, 413–417
4 York, P. (1983) Solid-state properties of powders in the formulation and 32 Bauer-Brandl, A. (1996) Polymorphic transitions of cimetidine during
processing of solid dosage forms. Int. J. Pharm. 14, 1–28 manufacture of solid dosage forms. Int. J. Pharm. 149, 195–206
5 Carstensen, J.T. (1988) Effect of moisture on the stability of solid dosage 33 Morris, K.R. et al. (1994) Characterization of humidity-dependent
forms. Drug Dev. Ind. Pharm. 14, 1927–1969 changes in crystal properties of a new HMG-CoA reductase inhibitor in
6 Morris, K.R. et al. (2001) Theoretical approaches to physical support of dosage form development. Int. J. Pharm. 108, 195–206
transformations of active pharmaceutical ingredients during 34 Airaksinen, S. et al. (2003) Effects of excipients on hydrate formation in
manufacturing processes. Adv. Drug Deliv. Rev. 48, 91–114 wet masses containing theophylline. J. Pharm. Sci. 92, 516–528
7 Brittain, H.G. and Fiese, E.F. (1999) Effects of pharmaceutical processing 35 Phadnis, N.V. and Suryanarayanan, R. (1997) Polymorphism in
on drug polymorphs and solvates. In Polymorphism in Pharmaceutical anhydrous theophylline: implications on the dissolution rate of
Solids, pp. 331-361, Marcel Dekker. theophylline tablets. J. Pharm. Sci. 86, 1256–1263
8 Brittain, H.G. et al. (1991) Physical characterization of pharmaceutical 36 Brittain, H.G. et al. (1993) Solid-state NMR and IR for the analysis of
solids. Pharm. Res. 8, 963–973 pharmaceutical solids: polymorphs of fosinopril sodium. J. Pharm.
9 Threlfall, T.L. (1995) Analysis of organic polymorphs, a review. Analyst Biomed. Anal. 11, 1063–1069
120, 2435–2460 37 Lefebvre, C. et al. (1986) Polymorphic transitions of carbamazepime
10 Rodriguez, N. and Morris, K.M. (1993) Hydrates. In Encyclopedia of during grinding and compression. Drug Dev. Ind. Pharm. 12, 1913–1927
Pharmaceutical Technology (Vol. 7), pp. 393–440, Marcel Dekker 38 Chan, H.K. and Doelker, E. (1985) Polymorphic transformation of some
11 Brittain, H. (ed.) (1995) Physical Characterization of Pharmaceutical Solids, drugs under compression. Drug Dev. Ind. Pharm. 11, 315–332
Marcel Dekker 39 Pirttimaki, J. et al. (1993) Effects of grinding and compression on crystal
12 Foster, B. (2003) Raman microscopy: taking chemical microscopy to the structure of anhydrous caffeine. Int. J. Pharm. 95, 93–99
next level. Am. Lab. April, 18–23 40 Otsuka, M. et al. (1989) Effects of mechanical energy of multi-tableting
13 Koheler, F. et al. (2002) NIR microscopy. Spect. Europe 14, 12 compression on the polymorphic transformations of chlorpropamide.
14 Katon, J.E. and Somer, A.J. (1992) IR microspectroscopy. Anal. Chem. J. Pharm. Pharmacol. 41, 665–669
64, 931–940 41 Matsumoto, T. et al. (1991) Effects of temperature and pressure during
15 Q2B Validation of analytical procedures: methodology (1997) Fed. Reg. compression on polymorphic transformation and crushing strength of
62, 27464 chlorpropamide tablets. J. Pharm. Pharmacol. 43, 74–78
16 Suryanarayanan, R. and Herman, C.S. (1991) Quantitative analysis of 42 Otsuka, M. and Matsuda, Y. (1993) Effects of environmental
the active tablet ingredient by powder X-ray difractometry. Pharm. Res. temperature and compression energy on polymorphic transformation
8, 393–399 during tabletting. Drug Dev. Ind. Pharm. 19, 2241–2269
17 Phadnis, N.V. et al. (1997) Identification of drugs in pharmaceutical dosage 43 Otsuka, M. et al. (1991) Effects of tableting pressure on hydration kinetics
forms by X-ray powder diffractometry. J. Pharm. Biomed. Anal. 15, 929–943 of theophylline anhydrate tablets. J. Pharm. Pharmacol. 43, 226–231
18 Stephens, P.W. (2002) What’s in your pill? What about theirs? Modern 44 Ando, H. et al. (1992) Effect of moisture on crystallization of
X-ray analysis in the pharmaceutical industry using synchrotron theophylline tablets. Drug Dev. Ind. Pharm. 18, 453–467
radiation. Am. Pharm. Rev. 5, 62–66 45 Hirayama, F. et al. (1994) Effect of 2-hydroxypropyl-β-cyclodextrin on
19 Stephenson, G.A. and Young, R. (2001) Potential applications of Rietveld crystallization and polymorphic transition on nifedipine in solid state.
analysis in the pharmaceutical industry. Am. Pharm. Rev. 4, 46–51 Pharm. Res. 11, 1766–1770
20 Chen, X. et al. (2001) Quantifying amorphous content of lactose using 46 Clarke, M.J. et al. (2000) Physicochemical factors governing the
parallel beam X-ray powder diffraction and whole pattern fitting. performance of nedocromil sodium as a dry powder aerosol. J. Pharm.
J. Pharm. Biomed. Anal. 26, 63–72 Sci. 89, 1160–1169
21 Smith, D.K. (1992) Particle statistics and whole pattern methods in 47 Miller, R.W. (2002) Process analytical technology (PAT) Part 1.
quantitative X-ray powder diffraction analysis. In Advances in X-ray Am. Pharm. Rev. 5, 25–29
Analysis (Vol.35), pp. 1–15 48 Miller, R.W. (2003) Process analytical technology (PAT) Part 2.
22 Mateos, A.G. (1992) Quantitative XRD analysis by partial least squares Am. Pharm. Rev. 6, 52–61
application in a commercial product. In Advances in X-ray Analysis 49 Cotton, M.L. et al. (1987) Drug-excipient interactioni study of enalapril
(vol. 35), pp. 117–126 maleate using thermal analysis and scanning electron microscopy.
23 Dash, A.K. et al. (2002) X-ray powder diffractometric method for Int. J. Pharm. 40, 129–142
quantitation of crystalline drug in microparticulate systems. I. 50 Guo, Y. et al. (2000) Physical characteristics and chemical degradation
microspheres. J. Pharm. Sci. 91, 983–990 of amorphous quinapril hydrochloride. J. Pharm. Sci. 89, 128–143
24 Saindon, P.J. et al. (1993) Solid-state nuclear magnetic resonance (NMR) 51 Byrn, S.R. et al. (2001) Chemical Reactivity in solid-state pharmaceuticals:
spectra of pharmaceutical dosage forms. Pharm. Res. 10, 197–203 formulation indications. Adv. Drug Deliv. Rev. 48, 115–136

www.drugdiscoverytoday.com 905

You might also like