You are on page 1of 8

Current Pharmaceutical Biotechnology, 2005, 6, 215-222 215

Application of Solution Calorimetry in Pharmaceutical and


Biopharmaceutical Research

P.G. Royall1,* and S. Gaisford2

1
Department of Pharmacy, Kings College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH
and 2Department of Pharmaceutics, School of Pharmacy, University of London, 29/39 Brunswick Square, London,
WC1N 1AX, UK

Abstract: In solution calorimetry the heat of solution (solH) is recorded as a solute (usually a solid) dissolves in an
excess of solvent. Such measurements are valuable during all the phases of pharmaceutical formulation and the number of
applications of the technique is growing. For instance, solution calorimetry is extremely useful during preformulation for
the detection and quantification of polymorphs, degrees of crystallinity and percent amorphous content; knowledge of all
of these parameters is essential in order to exert control over the manufacture and subsequent performance of a solid
pharmaceutical. Careful experimental design and data interpretation also allows the measurement of the enthalpy of
transfer (transH) of a solute between two phases. Because solution calorimetry does not require optically transparent
solutions, and can be used to study cloudy or turbid solutions or suspensions directly, measurement of transH affords the
opportunity to study the partitioning of drugs into, and across, biological membranes. It also allows the in-situ study of
cellular systems. Furthermore, novel experimental methodologies have led to the increasing use of solution calorimetry to
study a wider range of phenomena, such as the precipitation of drugs from supersaturated solutions or the formation of
liposomes from phospholipid films. It is the purpose of this review to discuss some of these applications, in the context of
pharmaceutical formulation and preformulation, and highlight some of the potential future areas where solution
calorimetry might find applications.
Key Words: Solution calorimetry, polymorphism, amorphous content, pharmaceuticals, dissolution, enthalpy of transfer.

1. INTRODUCTION Moreover, over time all the metastable polymorphs will


convert to the stable polymorph, which may have a
Many active pharmaceutical ingredients (APIs) exist in
disastrous effect on the efficacy of the formulated product. A
the solid-state or are formulated into solid dosage forms.
classic example of this is provided by chloramphenicol
This confers several advantages; stability in the solid-state is palmitate, which has three polymorphs, termed A, B and C
usually greater than in the liquid state, solids are easier to [1]. The A polymorph is unstable and is not allowed to be
transport, package and store and the majority of medicines,
included in any medicine; similarly, the C polymorph has
at least in the United Kingdom, are formulated as tablets as
virtually no appreciable bioavailability and its percentage in
the oral route is both quick and convenient. However, solid-
the final product must be strictly controlled. Any formulator
state formulations are usually slower-acting than liquid
of chloramphenicol palmitate must therefore know, and be
medicines containing the same active, because the first event
able to quantify, the amount of the B polymorph in any
that must occur following administration is that the API must
formulation.
dissolve in a suitable biological fluid (such as saliva or
gastric juices). On a molecular level, this requires the In addition to polymorphism, solid APIs may also exhibit
intramolecular forces holding the solid lattice together to be amorphicity (a lack of long-range molecular order). Because
overcome (endothermic) and the formation of new inter- amorphous materials dont have a lattice energy and are
actions with the solvent (generally exothermic). This event is essentially unstable (over time they will recrystallise to a
(usually) rate-limiting, and thus controls the observed crystalline form) they usually have appreciably faster
dissolution rate of the API from the formulation. dissolution rates than their crystalline equivalents, which
makes them especially suited for formulation into fast-acting
The intra-molecular forces in a crystalline material (the
medicines . The accidental inclusion of amorphous content
lattice energy) will vary if the material exhibits polymor- in what is otherwise presupposed to be a crystalline material
phism, (i.e. if the molecules can pack in more than one is also a hazard associated with processing of solid-state
crystalline formation), the most stable polymorph having the
highest lattice energy. It is therefore imperative that the
crystal form of any solid-state API is known, since the
dissolution rates of each polymorph will be different.
It is often stated that amorphous materials have higher solubilities than their
crystalline counterparts; since solubility is an equilibrium state, this cannot be true.
What may often happen is that the fast dissolution rate of the amorphous form results
*Address correspondence to this author at the Department of Pharmacy, in the production of a super-saturated solution which over time, although not
Kings College London, Franklin-Wilkins Building, 150 Stamford Street, necessarily in the time frame involved in drug administration and absorption, will
London, SE1 9NH, UK; Tel: +44 (0)20 7848 4780; Fax: +44 (0)20 7848 precipitate to form a saturated solution. As such, it should be stated that amorphous
materials may exhibit higher apparent solubilities than their crystalline equivalents.
4800; E-mail: paul.royall@kcl.ac.uk

1389-2010/05 $50.00+.00 2005 Bentham Science Publishers Ltd.


216 Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 Royall and Gaisford

materials (during, say, milling or compression steps) [2-3]. experiment, vary considerably (typically, semi-adiabatic
Although the percentage of amorphous content introduced in instruments are less sensitive and require much larger sample
this way is usually low (of the order of 1% w/w) its location volumes).
primarily on the surface of what are usually small particles
In a carefully constructed experiment, the enthalpy of
gives it a disproportionate control on the properties of the
solution, solH, is measured directly. Care must be taken to
material [4]. It is clear, then, that the detection and quanti- ensure that other contributing heat sources are accounted for.
fication of (often small) amorphous contents is of the utmost These may include effects from breaking the ampoule and
importance during the characterization of a solid pharma- sample stirring (most easily compensated for by subtracting
ceutical. a blank experiment from the sample experiment), changes in
One technique that offers the sensitivity to differentiate the solvent activity because of solute dissolution, changes in
between polymorphs as well as to detect low (typically the rate of evaporation of solvent into the headspace and
around 1% w/w) amorphous contents is solution calorimetry. changes in volume upon mixing of the two phases. The last
The principle of solution calorimetry is simple; the heat three effects can be assumed to be negligible if an ideal
change when a small quantity of a solid or liquid sample is (dilute) solution is formed; if this is not the case, a fact that
dispersed into a (relatively) large volume of solvent is would be confirmed by obtaining different values of solH
measured (either directly or by measuring a temperature with different amounts of sample, then the data must be
change which is subsequently converted to a heat change). extrapolated back to infinite dilution to give solH.
For a pure material dissolving into a solvent this results in
the enthalpy of solution (solH), an enthalpy change that 2.1. Types of Solution Calorimeter
reflects contributions from the bonds broken when the
Semi-adiabatic and heat-conduction instruments operate
crystal lattice breaks apart (latticeH) and from the bonds
on different principles and, while these have been discussed
formed when the molecules are solvated (solvationH). This
in detail elsewhere (See for example, 5-7) it is worth briefly
can be expressed as;
considering these differences here because they impact upon
sol H = lattice H + solvation H Equation 1 the subsequent discussion of the data.
Depending on the relative magnitudes of latticeH and 2.1.1. Semi-Adiabatic Solution Calorimeters
solvationH, the heat of solution can have a positive (endo- In an ideal adiabatic calorimeter there is no heat ex-
thermic) or negative (exothermic) sign. Careful experimental change between the calorimetric vessel and its surroundings.
design (discussed in more detail below) is required to ensure This is usually attained by placing an adiabatic shield around
that the measured enthalpy does not contain erroneous the vessel. Thus any change in the heat content of a sample
contributions from other sources, such as dilution (liquid as it reacts causes either a temperature rise (exothermic
samples), friction effects (ampoule breaking and sample processes) or fall (endothermic processes) in the vessel. The
stirring) and sample-sample interactions (if an ideal solution change in heat is then equal to the product of the temperature
is not formed), or simply as a corollary of poor experimental change and an experimentally determined proportionality
design or execution. Since solH differs for each polymorph constant (or calibration constant, ). The proportionality
of a solid sample, it is clear that solution calorimetry constant is usually determined by electrical calibration.
provides valuable information that allows the detection and Thus;
quantification of polymorph content.
q
Moreover, since solH contains information on the attrac- T = Equation 2
tive forces holding a solid together, solution calorimetry can
provide fundamental information on the nature of multi- dT
component systems. Such systems include solid-dispersions, = Equation 3
dt
polymeric systems and proteins freeze-dried with carbo-
hydrates. As such, solution calorimetry is a powerful tool where represents power. Ideally, the value of returned
that can be used to investigate a wide range of pharma- after calibration should equal the heat capacity of the
ceutical systems, and it is the purpose of this review to calorimeter vessel (Cv, the vessel including the calorimetric
discuss some of these applications in the context of the ampoule, block, heaters, thermopiles and the sample) but in
preparation and formulation of novel pharmaceuticals and practice losses in heat mean the value may differ slightly.
biopharmaceuticals. However, assuming the losses are the same for both sample
and reference, the power value returned will be accurate. It is
2. THE PRINCIPLES OF SOLUTION CALORIMETRY also the case that Cv varies depending on the heat capacity of
the sample being studied, which affects the measuring
A solution calorimeter records the heat change when a sensitivity of the instrument. Thus, a small heat capacity
sample (solid or liquid) is dissolved in a large volume of results in a large rise in temperature for a given quantity of
solvent. There are two types of solution calorimeter design heat and, consequently, better sensitivity. However, calori-
commercially available; instruments that operate on a semi- meters with low heat capacities are more sensitive to
adiabatic principle (i.e. that record a temperature change environmental temperature fluctuations and have lower
upon reaction) and instruments that operate on a heat- baseline stabilities. Any calorimeter design therefore results
conduction principle (i.e. that record a power change directly in a compromise between baseline stability and measurement
upon reaction). The sensitivities of these instruments, and sensitivity.
hence the quantities of solute and solvent required for
Application of Solution Calorimetry in Pharmaceutical Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 217

In practice, true adiabatic conditions are difficult to


achieve and there is usually some heat-leak to the surround- = Equation 6
k
ings. If this heat-leak is designed into the calorimeter (such
as is the case with the SolCal, an example of a commercial where k is the heat exchange coefficient of the vessel.
semi-adiabatic calorimeter, Thermometric AB, Jrflla, The values of T and are calculated by analysis of the
Sweden) the system operates under semi-adiabatic (or baseline regions immediately preceding and following the
isoperibol) conditions and corrections must be made in order break. These baseline sections will be approaching the
to return accurate data. These corrections are usually based temperature of the surrounding heat-sink exponentially and
on Newtons law of cooling (the most common being the are described by;
method of Regnault-Pfaundler, discussed below). t
In the case of the SolCal (the principles apply to all T = T + (T0 T )e Equation 7
similar solution calorimeters), at the start of an experiment
The data are then fitted to Equation 7 using a least-
the instrument is held above or below the temperature of its
squares minimising routine to return values for T and .
thermostatting bath (typically by up to 200 mK). With time
the instrument will approach the temperature of the Once these are known, Tadj can be calculated. This value is
thermostatting bath; data capture is initiated when this then used to calculate Tcorr for the sample break and also for
approach becomes exponential (this assumption is a the two electrical calibrations (Tcorr, calibration). There should
be no significant difference in the Tcorr, calibration values
necessary precursor to employing the heat-balance equations
used to calculate the heat evolved or absorbed by the system determined for the two calibrations and an averaged value is
used. The calibration constant is then determined from;
contained within the vessel). Thus, the response due to
dissolution, and any electrical calibrations (usually two are Qcalibration
performed; one before and one after the break to ensure the = Equation 8
heat capacity of the system has remained constant), must be Tcorr,calibration
performed before the instrument reaches thermal equilibrium the heat-change for the break is then easily determined;
with the bath. In practice, this limits the technique to
studying events that, ideally, reach completion in less than Qreaction = .Tcorr Equation 9
30 min.
2.1.2. Heat-Conduction Calorimeters
Upon completion of an event in a solution calorimeter a
quantity of heat will have been recorded. As noted above, the A heat-conduction calorimeter is surrounded by a heat-
heat will be given by the product of the temperature change sink, which acts to maintain the system at a constant tem-
and the calibration constant (which in the ideal case is the perature. Between the vessel and the heat-sink is a thermo-
heat capacity of the vessel). This interpretation assumes that pile wall. Any heat released or absorbed upon reaction is
the measured temperature change arises solely from the quantitatively exchanged with the heat-sink. The thermopiles
event occurring in the vessel; in practice, other events, such generate a voltage signal that is proportional to the power
as ampoule breaking, stirring and heat-leakage, all contribute flowing across them; this signal is amplified, multiplied by
to the temperature change of the vessel. For accurate data the cell constant (determined through electrical calibration)
analysis these effects must be removed from the observed and recorded as power versus time. An isothermal system is
temperature change (Tobs) to give Tcorr, the temperature not limited to reaction processes that reach completion
change that would have occurred under ideal conditions. within 30 min, as semi-adiabatic instruments are, because it
Thus; is always (essentially) in equilibrium with its surrounding
heat-sink. Furthermore, the greater measuring sensitivity of
Tobs = Tcorr + Tadj Equation 4 the thermopiles (as opposed to the thermisters used in semi-
adiabatic instruments) means that smaller sample masses can
where Tadj is defined as the temperature change arising be used.
from all the other contributing events in the vessel. Usually
the method of Regnault-Pfaundler, which is based on the A further consideration of the use of isothermal
dynamics of the break, is used to determine the value of Tadj instruments concerns dynamic correction of the data. The
[8]. In this case; aim of dynamic correction is to remove the effect of the
thermal inertia inherent in any calorimeter (i.e. the delay
tend between heat being released by the sample and that heat
1
Tadj = (T T)dt Equation 5 causing a measurable voltage to be generated by the
tstart thermopiles) and it is principally used for short-term events,
typically in titration experiments. However, since one
where T is the temperature of the vessel and its contents at outcome from dynamic correction is an improvement in the
time t, T is the temperature that the vessel would attain after signal to noise (S/N) ratio of the data it offers the potential to
an infinitely long time period, tstart and tend are the start and reduce the standard deviation of dissolution experiments,
end times of the experiment respectively and is the time because peak areas can be determined with greater precision.
constant of the instrument. Note that T is effectively the
value of T at t and is commonly described as the steady- In the case of a typical instrument (the 20 ml micro
state temperature of the vessel. The time constant has units reaction ampoule, Thermometric AB, Sweden), dynamic
of seconds and can also be expressed as; correction is achieved by application of a modified form of
the Tian equation;
218 Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 Royall and Gaisford

dPR d2P standards offer a better alternative and have been the subject
PC = PR + ( 1 + 2 ). + 1 . 2 . 2R Equation 10 of some discussion in the literature.
dt dt
There are a number of requirements imposed upon a test
where PR and PC are the raw and corrected powers
reaction; it should be robust, simple to perform, require
respectively and 1 and 2 are termed the first and second
commonly available materials that require no special
time constants of the instrument (for a further discussion of preparation prior to use and it should be applicable across a
the derivation and use of Equation 1 see, for example, [9]). range of instrumentation [10]. A number of chemical test
The time constants, 1 and 2, are determined by a least reactions for solutions calorimeters have been proposed and
squares analysis of data following an electrical calibration discussed, including the dissolution of Tris in 0.1 M HCl [5,
(and are hence not user defined). It is important to note that a
11], the dissolution of KCl or NaCl in water [12-14] and the
number of assumptions are made in order to derive Equation
dissolution of propan-1-ol in water [15]. The dissolution of
10 (the major assumption being that there are no temperature
sucrose in water can also be used [16-17], although this is
gradients within the sample) and that it only approximates
not currently recognised as a test reaction.
the true dynamic delay inherent to the instrument. The
corrected data so produced, while much more closely Of these systems, the dissolution of KCl into water is
resembling the true response of the sample, therefore often usually recommended because it is robust, easy to perform
contain artefacts, such as overshoots where both endo- and and a standard reference material is available from NIST (the
exothermic events are indicated even though it is known that National Institute for Standards and Technology, USA); the
only one event is occurring in the sample. In principle, these enthalpy of solution of the NIST certified KCl into water is
artefacts could be removed by altering the values of 1 and 2 17.584 0.017 kJ mol -1 [13]. However, the use of KCl is not
but this is difficult in practice. It is therefore easier to use without drawbacks; principally, the value of solH varies as a
corrected data to determine reaction enthalpies only and to function of the concentration achieved after dissolution,
note that the use of such data to elucidate kinetic information because of the effect of the enthalpy of dilution (L); thus,
must be undertaken with caution. It can be shown that the the certified value for the NIST reference material of 17.584
total net heat change recorded for both dynamically kJ mol-1 applies only if a final concentration of 0.111 mol kg-
1
corrected and raw data, in the ideal case, is the same [9]. is attained in the calorimetric vessel. This corresponds to a
molar ratio of water to KCl of 500 to 1 and is often written
2.1.3. Sample Solvent Mixing as solH (500 H2O, 298.15 K). If measurements are per-
There are several methods by which the solute and formed under different conditions, then the value obtained
solvent can be introduced. The solute and solvent can be (nH2O, 298.15 K) must be corrected to that which would
held in separate chambers of the same cell - rotating the have been recorded at 500 H2O, in order to draw com-
sample cell end-over-end mixes the materials and initiates parison. These corrections are explained in the certification
the interaction (this is the system employed by, for instance, certificate supplied with the NIST sample [13], although the
the C80, Setaram, France); the solute can be held in a sealed data supplied there apply only to experiments performed
glass ampoule which is broken into the solvent to initiate where n varies from 100 to 1000.
reaction (used in the 2225 precision solution calorimeter, The effects of KCl concentration on solH have been
Thermometric AB, Sweden); the solute can be held in studied extensively by Kilday [12], who corrected the
reusable metal canisters which are broken into the solvent observed enthalpy values over a range of water ratios (n =
(used in the 20 ml micro solution ampoule, Thermometric 500 to 10000) to account for the enthalpy of dilution; this
AB, Sweden); or liquid solutes/solutions can be titrated into resulted in a value for solH, the enthalpy of solution at
a cell containing solvent (such as the VP-ITC, Microcal Inc., infinite dilution (solH = 17.241 0.018 kJ mol-1).
USA).
Modern solution calorimeters often use microgram
2.2. Calibration samples and are capable of detecting very small powers; one
consequence of this is that it is not possible to perform the
Calibration is vital to ensure that instruments are opera- KCl experiment under the NIST certification conditions
ted properly and are functioning correctly, that data from because the heat generated would be of a magnitude
different instruments or different laboratories are comparable sufficient to saturate the amplifiers. Because of this several
and, perhaps most importantly when considering pharma- recent studies have been conducted looking at the applicabi-
ceutical samples, that the data obtained are validated and can lity of test reactions to modern solution calorimeters [7, 18].
be incorporated into regulatory documents. Most calori- The outcome from these studies appears to be that if KCl is
meters are calibrated using an electrical substitution method. to be used then it is a smaller, and easier, correction to the
In this case, a resistance heater (usually located under the enthalpy of solution at infinite dilution, but that sucrose may
sample cell) produces a known amount of heat, which is offer a better, and cheaper, alternative, especially for heat-
measured by the instrument. The heat output recorded is conduction instruments that use very small (mg) samples.
adjusted until the measured and expected heats are the same
(by multiplying the raw data signal by a constant value; the 3. APPLICATIONS
cell constant). However, it is debatable whether an electrical
calibration truly represents an accurate method by which to 3.1. Polymorphism
validate instrument performance, primarily because the As mentioned above, if a drug is prepared in the solid-
source and rate of heat generation will be different from that state then it is highly likely it will exhibit polymorphism and
caused by a chemical interaction. In this sense, chemical it is imperative that the polymorph(s) present in a sample are
Application of Solution Calorimetry in Pharmaceutical Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 219

known before formulation, such that the resulting medicine of forms I, II and III respectively. It was determined that
performs within its pharmacopoeial specifications. For new HIII equaled 22.98 kJ mol-1.
drug entities, the pressure to bring a product to market
Since Hesss law allows the interpretation of solH by
usually means that the developing company doesnt have the
invoking any number of steps, as long as the starting state is
time to characterise the relative bioavailability of each
the solid and the final state is the solution, one approach to
polymorphic form; usually, the most stable form is selected
for development and the formulation is tailored to ensure its the analysis of dissolution data is to consider the heat as
stability. This formulation strategy ensures compliance with comprising two major contributions; one which represents
the guidance provided by the International Commission on the breaking of the solid-state bonds and the other which
Harmonisation (ICH) on the selection of solid forms of drugs represents all the processes through which the molecules
become solvated [26]. If bond breaking is considered to be
[19]. Similarly, for Abbreviated New Drug Applications
similar to a melting process, then it can be represented by
(ANDA) the sponsor must provide evidence that the pro-
posed generic product and the original product are pharma- fH298, the enthalpy of formation of a supercooled liquid at
ceutically equivalent and bioequivalent [20]; clearly, not 298 K (assuming the dissolution experiment is conducted at
having any control or knowledge of the polymorphic forms 298 K), while the enthalpy changes associated with mixing
present would render submission of such data impossible. and solvation can be represented by m H. This can be
represented as shown in Fig. 1.
Solution calorimetry allows the direct measurement of
the lattice energy of a sample, which alters with each poly-
morphic form of the material and, although other analytical
techniques may be used to discriminate between polymorphs
(such as DSC, XRPD, solid-state NMR, IR and Raman
spectroscopy and microscopy [21]), the technique has three
principle advantages; firstly, the measurement is performed
directly at the storage (or, indeed, any desired) temperature,
in contrast to DSC measurements which usually need to be
extrapolated to give an indication of structure at room
temperature; secondly, the data allow comparison between
samples, giving insights into batch-to-batch or formulation
variability; and thirdly the various contributions to the
measured heat of solution can be dissected out by careful
experimentation, allowing a mechanistic analysis of the Fig. (1). A description of dissolution, based on Hesss law [26].
process under investigation. The principal drawback, related
to the latter point above, is that the heat of solution is a This model has been applied to the study of the dissolu-
composite of all those processes that are occurring during tion of various poly(ethylene) glycols, commonly encoun-
dissolution and it may sometimes be impossible to separate tered pharmaceutical excipients, in water [26], where fH298
the heat into its component parts. was measured using DSC data and solH was measured in
Solution calorimetry has been used to assess the different the solution calorimeter. Four different molecular weight
polymorphs of many drugs. For instance, forms I and II of PEGs were selected (3, 400, 6000, 10, 000 and 20, 000) and
cyclopenthiazide have been shown to have comparable samples were prepared with different thermal histories;
enthalpies of solution (~6 kJ mol-1) while form III has a samples were quench-cooled from the melt, slow-cooled
higher enthalpy of solution of 15 kJ mol-1 [22]. Similarly, from the melt (5 oC min-1) or left untreated. It was found that
solution calorimetry has been used to characterise the three solH varied as a function of both molecular weight and
polymorphic forms of a pre-clinical drug, Abbott-79175 [23] thermal history, although no discernable relationships could
and the polymorphs of an angiotensin II antagonist agent be found. According to the model above, mH should be
(MK996) [24]. constant for each molecular weight PEG, independent of any
thermal history, since this represents the dissolution of
A study of urapidil resulted in the enthalpies of solution individual molecules, while fH298 should be highly depen-
of forms I and II of the drug (21.96 and 23.89 kJ mol-1 dent on thermal history. Accordingly, the soHl values will
respectively) being determined [25]. However, the authors vary, since this parameter is the sum of fH298 and mH.
note that urapidil exists in an additional form (form III), Subtraction of the fH298 values, determined from DSC data,
which they were unable to obtain in a pure form. By mea- from the measured solH values showed that, within a 5%
suring the heat of solution of the mixture (mixH), and using confidence level the mH values did remain constant for
DSC data to determine the percentage of each polymorph in each molecular weight PEG, and that the measured
a sample containing form III (7.4% form I, 2.7% form II and variabilities in the measured heats of solution derived from
89.9% form III), they calculated the enthalpy of solution of changes in fH298.
form III using the following relationship;
The correlation of the heats of solution of different
mix H = ( X I .H I ) + ( X II .H II ) + ( X III .H III ) Equation 11 polymorphs of a drug with dissolution rates has also been
attempted. For instance, Terada et al. [27] showed there was
where XI, XII and XIII are the fractions of form I, II and III a linear correlation between the heats of solution of different
respectively and HI, HII and HIII are the heats of solution polymorphs of indomethacin and the logarithms of their
220 Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 Royall and Gaisford

initial dissolution rates (determined by the rotating disk many solid drugs are prepared in the amorphous state
method). The same authors showed a similar relationship for although, as noted earlier, knowledge of the percent amor-
a range of samples of terfenadine of varying crystallinity. phous content and its stability are vital prerequisites to
ensure the stability and continued efficacy of the medicine
In some cases, a drug will be insoluble in water, and in upon storage.
order to derive some meaningful information on its poly-
morphic forms it is desirable to measure heats of solution in The use of solution calorimetry to quantify the degree of
two non-aqueous solvents. Again, in accordance with the crystallinity in a solid sample is predicated on the
model discussed above, the value of solH for each poly- relationship shown in Equation 11, where the measured heat
morph will alter in each solvent. However, the difference of solution is given by the sum of the enthalpies and weight
between the heats of solution for each polymorph should be fractions for the crystalline and amorphous states present.
constant irrespective of the solvent used; The usual methodology is to prepare a calibration line of
solH against degree of crystallinity using a number of
sol, form I H sol, form II H = T H = Constant Equation 12 known standards (usually prepared by blending the
appropriate mass quantities of wholly amorphous and wholly
where TH is the transfer enthalpy, and is the energy crystalline material); the calibration plot should be linear and
required to transfer between the two forms. Differences in can thus be used to determine the degree of crystallinity in
solH determined in each solvent will reflect changes in the an unknown sample.
solvation interaction with that solvent. There are two potential drawbacks to using this approach.
This approach has been used to investigate the poly- Firstly, the sample may exhibit polymorphism, in which case
morphs of enalapril maleate, by recording the heats of there may be more than one crystalline form present; if this
solution of forms I and II of the drug in acetone and metha- is the case then, for the calibration plot to be linear, it must
nol [28]. The differences in the heats of solution of forms I be ensured that the crystalline material used to prepare the
and II in each solvent (2.88 and 2.13 kJ mol-1 in acetone and standards and the unknown sample contain the same
methanol respectively) were the same within experimental proportions of the polymorphs. Secondly, it is likely that the
error, and gave TH, the energy that would be required for interactions in a particle that has a crystalline core and
form II to convert to form I. The difference in the heat of amorphous material on its surface (the likely situation for a
solution for forms I and II in methanol, compared with processed pharmaceutical) differ from those of wholly
acetone, was 23.53 kJ mol-1; that is, the heat of solution in amorphous and wholly crystalline particles, which may
methanol was more exothermic than in acetone. This value result in the calibration plot producing spurious results.
was found to be consistent with the formation of a single The first use of solution calorimetry for the quantitative
hydrogen bond. measurement of the degree of crystallinity of pharma-
Similar data, recorded using ethanol and methanol, have ceuticals was by Pikal et al. [31], who measured the heat of
been recorded for terfenadine [29], where a difference in solution of various -lactam antibiotics. Subsequent studies
energy of approximately 13 kJ mol-1 was found between include the analysis of sulphamethoxazole from different
forms I and II. In this case, it was observed that solH for any sources [32], sucrose [16] and clathrate warfarin sodium
solid form of terfenadine was around 4 kJ mol-1 lower in [16].
methanol than ethanol, indicating that polar groups on the In many of these studies, the aim was to quantify relat-
drug molecule play an important role in the solute/solvent ively large mass percentages of crystalline material, giving
interaction. heat changes easily within the detection limit of the tech-
A different approach to study water insoluble drugs is to nique. The issue of detection limits becomes more important
use a concentrated surfactant solution instead of pure water if the objective of the study is to assess the quantity of
as the solvent. For instance, the enthalpies of solution of a amorphous material present in what is a predominately
number of cimetidine polymorphs have been studied using crystalline sample because, as stated earlier, in a milled
concentrated SDS and Tween 20 solutions [30]. sample amorphous material may typically only be present up
to 1% w/w. An assessment of the applicability of solution
3.2. Determination of Degree of Crystallinity/Amorphous calorimetry to study small amorphous contents in solid
Content pharmaceuticals was conducted by Hogan and Buckton [4],
who prepared a calibration curve for lactose between 0 and
Solid pharmaceuticals may often not be entirely 10% w/w amorphous content in the same way as described
crystalline and contain regions of amorphous content. An above. They found that the technique could quantify
amorphous material (most simply described as a material amorphous content to 0.5% w/w but noted that care needed
with the structure of a liquid and the viscosity of a solid) is to be taken when preparing the ampoules, because ingress of
characterised by a lack of any long-range crystal structure even small amounts of humidity caused partial recrystalli-
and, because it therefore has no lattice energy, the lack of a sation of the sample before measurement.
melting point (upon heating, an amorphous solid will
recrystallise and it is the crystalline form that subsequently Usually, in experiments designed to measure degrees of
melts). Amorphous solids are thermodynamically unstable crystallinity of amorphous content a solvent is selected in
and usually exhibit fast dissolution rates compared with their which the solute is freely soluble. This ensures complete
crystalline equivalents. Pharmaceutically, this is a big advan- dissolution of the sample within the time frame of the
tage for medicines that are required to be fast acting and experiment. Harjunen et al. [33] studied the dissolution of
Application of Solution Calorimetry in Pharmaceutical Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 221

lactose into saturated aqueous lactose solutions, a system 3.4. Other Applications
where clearly the solute would not completely dissolve.
Interestingly, they observed a linear relationship between the Perlovich and Bauer-Brandl [42] have discussed the use
amorphous content of the lactose solute and the measured of heat of solution data to predict drug solubility using two
heat of solution in the saturated lactose solution (satH). model compounds, benzoic acid and aspirin. Their data
suggest it may be possible to predict the solubility or
Similarly, a linear relationship was found between the amor-
phous content of lactose and satH in methanolic saturated solvation of a drug in different media. Similarly, Willson and
Sokoloski [43], as part of a study developing a method to
solutions of lactose [34].
rank the stability of drug polymorphs, correlated solution
3.3. Characterisation of Interactions calorimetry measurements to conventionally determined
solubility data. Solution calorimetry has also been employed
As discussed above, application of Hesss law allows the to study the dissolution and solvation of the model systems,
construction of solution calorimetry experiments that allow flurbiprofen and diflunisal [44] and to compare the solvation
valuable information on the interaction of a solvent with a of (+)-naproxen with three model NSAIDs (benzoic acid,
mixed solvent system. An example would be to measure the diflunisal and flurbiprofen) [45].
dissolution of a solid in a buffer and a buffered solution of
micelles. The difference in the solH values recorded in the Recent work has shown that solution calorimetry can be
two systems would represent the enthalpy of transfer of the used to investigate the stability of supersaturated systems
solute from the buffer to the micelle. Similarly, if a complex [46]. Supersaturated systems are particularly important for
solvent system is employed, such as one that matched a topical and transdermal formulations where the API is may
biological medium for instance, it would be possible to formulated above its solubility in order to maximise the
measure the enthalpy of interaction of a solute with the entire diffusional driving force for absorption [47, 48]. Clearly,
medium and then each of its constituents in turn. supersaturated formulations are inherently thermodynami-
cally unstable and it is hence likely that crystallisation of the
This approach has been used to investigate the interaction active will occur during storage with a resultant change in
of two solutes, propranolol HCl and mannitol, with two drug bioavailability. The ability to measure the time taken
simulated intestinal fluids (fasted-state and fed-state) and before recrystallisation occurs is therefore an essential
Hanks balanced salt solution (HBSS) [35]. The simulated prerequisite to the development of such transdermal formula-
intestinal fluids contained bile salts and lipids which formed tions and is difficult because the formulations are often
mixed micelles while no micelles formed in HBSS. It was opaque semi-solids.
found that the two solutes exhibited endothermic heats of
solution in all solvents; however, the values for propranolol Solution calorimetry allows the study of these formula-
HCl were lower in the two simulated fluids than in HBSS tions because it permits the in-situ formation of saturated
while the values for mannitol were constant in all media. solutions. Hadgraft et al. [46] have shown that by adding
Calculation of the enthalpy of transfer (transH) for pro- ibuprofen in a co-solvent to a saturated solution of ibuprofen
pranolol HCl into the micelles in the two simulated fluids it is possible to form a supersaturated drug solution in the
revealed an exothermic (and hence favourable) interaction calorimetric ampoule. In the absence of any stabilising com-
(-10.3 kJ mol-1 for the fed-state and -2.1 kJ mol-1 for the pound the supersaturated system immediately precipitates
fasted-state). and a heat signal is observed. If a small concentration (0.1%
w/v) of hydroxypropyl methyl cellulose (HPMC) is added to
In an earlier study, Beezer et al. [36] calculated values of
the system the time to recrystallisation increased to
transH for a series of alkoxyphenols to Escherichia coli cells,
approximately 8 minutes. At a concentration of HPMC of
while the combination of Caco-2 cells and simulated
0.5 % w/v the supersaturated system remained stable for
intestinal fluids has been suggested as a model for studying
approximately 30 minutes, while at an HPMC concentration
drug permeability through membranes [37]. It seems as if
of 1% w/v the system was stable for longer than 24 hours.
solution calorimetry may an ideal technique by which to
monitor these interactions, especially as it is unaffected by Barriocanal et al. [49] studied the formation of liposome
cloudy or turbid solutions or suspensions. formation by coating the inside of a glass ampoule with
phospholipids (deposited from a chloroform solution). Upon
Chada et al. [38] used solution calorimetry to probe the
interactions of diclofenac sodium in cyclodextrin solutions breaking, the phospholipids film hydrated and liposomes
formed; the solution calorimeter measured the changes in
and water/ethanol mixtures. Tong et al. [39] used solution
calorimetry to evaluate the stability constants and enthalpy heat associated with the processes. They found that the
formation of liposomes from egg phosphatidylcholine was
changes associated with the formation of complexes between
2-hydroxypropyl--cyclodextrin and a group of 12 amine exothermic while the formation of liposomes from dimyris-
toylphosphatidylcholine was endothermic and suggest that
compounds which all had a diphenylmethyl functional
group. They found that only terfenadine HCl formed a 1:2 this difference arose from the influence of the hydrocarbon
chains predominately on the hydration process. They also
complex with the -cyclodextrin, the other 11 compounds all
forming 1:1 complexes. noted that the retention of small quantities of chloroform in
the phospholipids film significantly altered the enthalpy
Solution calorimetry has also been used to measure the change of liposome formation, an effect that was ascribed to
enthalpy of solution of diclofenac sodium, paracetamol and the effect of chloroform on hydration.
their binary mixtures [40] and to evaluate the in vitro
compatibility of amoxicillin/clavulanic acid and ampicillin/ Solution calorimetry can also be used to study the actions
sulbactam with ciprofloxacin [41]. of formulations directly. For instance, Gaisford et al. [50]
222 Current Pharmaceutical Biotechnology, 2005, Vol. 6, No. 3 Royall and Gaisford

used solution calorimetry to follow the acid neutralisation of [14] Archer, D.G. and Kirklin, D.R. (2000) Thermochimica Acta, 347,
magnesium trisilicate mixture BP. They showed that of the 21-30.
[15] Olofsson, G.; Berling, D.; Markova, N. and Molund, M. (2000)
three active components in the mixture (magnesium carbo- Thermochimica Acta, 347, 31-36.
nate, magnesium trisilicate and sodium bicarbonate), magne- [16] Gao, D. and Rytting, J.H. (1997) Int. J. Pharm., 151, 183-192.
sium carbonate contributed most to the action of the product, [17] Salvetti, G., Tognoni, E., Tombari, E. and Johari, G.P. (1996)
followed by sodium bicarbonate. The results suggested that Thermochimica Acta, 285, 243-252.
[18] Ramos, R., Gaisford, S., Buckton, G., Royall, P.G., Yff, B.T.S. and
magnesium trisilicate did not neutralise a significant quantity ONeill, M.A.A. (2005) Int. J. Pharm., Submitted for publication.
of acid by itself but that it extended the neutralising response [19] Grant, D.J.W. and Byrn, S.R. (2004) Adv. Drug Del. Rev., 56, 237-
of the mixture, presumably by interacting with the 239.
magnesium carbonate. The results provided an insight into [20] Raw, A.S., Furness, M.S., Gill, D.S., Adams, R.C., Holcombe Jr,
the mechanism of action of the product and the data would F.O. and Yu, L.X. (2004) Adv. Drug Del. Rev., 56, 397-414.
[21] Yu, L., Reutzel, S.M. and Stephenson, G.A. (1998) Pharm. Sci.
be useful in any reformulation exercise. Tech. Today, 1, 118-127.
[22] Gerber, J.J.; vanderWatt, J.G. and Ltter, A.P. (1991) Int. J.
SUMMARY Pharm., 73, 137-145.
[23] Li, R.C.Y., Mayer, P.T., Trivedi, J.S. and Fort, J.J. (1996) J.
Solution calorimetry offers the same virtues to the Pharm. Sci., 85, 773-780.
pharmaceutical scientist as other forms of calorimetry; it is [24] Jahansouz, H., Thompson, K.C., Brenner, G.S. and Kaufman, M.J.
non-invasive, non-destructive and is not limited to the study (1999) Pharm. Dev. Tech., 4, 181-187.
[25] Botha, S.A., Guillory, J.K. and Lotter, A.P. (1986) J. Pharm.
of homogeneous systems. It does not require large quantities Biomed. Anal., 4, 573-587.
of sample and the data are easy to interpret, so long as care [26] Craig, D.Q.M. and Newton, J.M. (1991) Int. J. Pharm., 74, 43-48.
has been taken in experimental design and execution. Solu- [27] Terada, K., Kitano, H., Yoshihashi, Y. and Yonemochi, E. (2000)
tion calorimetry has classically been used during preformula- Pharm. Res., 17, 920-924.
[28] Ip, D.P.; Brenner, G.S.; Stevenson, J.M.; Lindenbaum, S.; Douglas,
tion, where it allows the detection and identification of A.W.; Klein, S.D. and McCauley, J.A. (1986) Int. J. Pharm., 28,
polymorphs and the quantification of crystalline or amor- 183-191.
phous contents. However, its ability to study cloudy or turbid [29] Canotilho, J.; Costa, F.S.; Sousa, A.T.; Redinha, J.S. and Leitao,
solutions or suspensions means it is finding increasing M.L.P. (1997) Thermochimica Acta, 299, 1-6.
applications in the direct study of the interactions of drugs [30] Souillac, P.O.; Dave, P. and Rytting, J.H. (2002) Int. J. Pharm.,
231, 185-196.
with complex biological systems, where it is possible to [31] Pikal, M.J., Lukes, A.L., Lang, J.E. and Gaines, K. (1978) J.
measure the enthalpy of transfer of a drug between phases. Pharm. Sci., 67, 767-772.
Novel experimental strategies are also expanding its range of [32] Guillory, J.K. and Erb, D.M. (1985) Pharm. Manuf., Sep, 28-33
applications, and it has been used to investigate the stability [33] Harjunen, P., Lehto, V-P., Koivisto, M., Levonen, E., Paronen, P.
and Jrvinen, K. (2004) Drug Dev. Ind. Pharm., 30, 809-815.
of supersaturated systems, antacid formulations and the [34] Katainen, E., Niemel, P., Pllysaho, M., Harjunen, P., Suhonen,
formation of liposomes. Its continued use and development J. and Jrvinen, K. (2003) Eur. J. Pharm. Sci. Abstracts, 19, S36.
will ensure it retains its place as a vital tool in pharmaceut- [35] Arnot, L.F., Minet, A., Patel, N., Royall, P.G. and Forbes, B.
ical formulation and preformulation. (2004) Thermochimica Acta, 419, 259-266.
[36] Beezer, A.E., Gooch, C.A., Hunter, W.H. and Volpe, P.L.O. (1987)
J. Pharm. Pharmacol., 39, 774-779.
REFERENCES [37] Patel, N., Murray, J.G. and Forbes, B. (2001) British
[1] Augiar, A.J., Krc Jr. J., Kinkel, A.W. and Samyn, J.C. (1967) J. Pharmaceutical Conference Abstracts Book, p51.
Pharm. Sci., 56, 847-853. [38] Chadha, R., Kashid, N., Kumar, A. and Jain, D.V.S. (2002) J.
[2] Grant, D.J.W. and York, P. (1986) Int. J. Pharm., 30, 161-180. Pharm. Pharmacol., 54, 481-486.
[3] Giron, D., Remy, P., Thomas, S. and Vilette, E. (1997) J. Thermal [39] Tong, W.Q., Lach, J.L., Chin, T.F. and Guillory, J.K. (1991) J.
Analysis, 48, 465-472. Pharm. Biomed. Anal., 9, 1139-1146.
[4] Hogan, S. and Buckton, G. (2000) Int. J. Pharm., 207, 57-64. [40] Chada, R., Kashid, N. and Jain, D.V.S. (2003) J. Pharm. Biomed.
[5] Hill, J.O.; Ojlund, G. and Wads, I. (1969) J. Chem. Thermodyn., Anal., 30, 1515-1522.
1, 111-116. [41] Chada, R., Kashid, N. and Jain, D.V.S. (2004) J. Pharm. Biomed.
[6] Bastos, M., Bai, G., Qvarnstrm, E. and Wads, I. (2003) Anal., 36, 295-307.
Thermochimica Acta, 405, 21-30. [42] Perlovich, G.L. and Bauer-Brandl, A. (2003) Pharm. Res., 20, 471-
[7] Yff, B.T.S.; Royall, P.G.; Brown, M.B. and Martin, G.P. (2004) 478.
Int. J. Pharm., 269, 361-372. [43] Willson, R.J. and Sokoloski, T.D. (2004) Thermochimica Acta,
[8] Wads I (1966) The LKB Inst J, 13, 33-39. 417, 239-243.
[9] Randzio, S.L. and Suurkuusk, J. (1980) In Beezer AE, editor. [44] Perlovich, G.L., Kurkov, S.V. and Bauer-Brandl, A. (2003) Eur. J.
Biological microcalorimetry. 1st ed., London:Academic Press Pharm. Sci., 19, 423-432.
p311-341. [45] Perlovich, G.L., Kurkov, S.V., Kinchin, A.N. and Bauer-Brandl, A.
[10] Beezer, A.E., Hills, A.K., ONeill, M.A.A., Morris, A.C., Kierstan, (2004) Eur. J. Pharm. Biopharm., 57, 411-420.
K.T.E., Deal, R.M., Waters, L.J., Hadgraft, J., Mitchell, J.C., [46] Hadgraft, J., ONeill, M.A.A., Gaisford, S., Beezer, A.E., Al-
Connor, J.A., Orchard, J.E., Willson, R.J., Hofelich, T.C., Beaudin, Moshey, L., Farah, F. and Auner, B. (2005) J. Drug Del. Sci. Tech.,
J., Wolf, G., Baitalow, F., Gaisford, S., Lane, R.A., Buckton, G., Submitted for publication.
Phipps, M.A., Winneke, R.A., Schmitt, E.A., Hansen, L.D., [47] Lervolino, M., Raghavan, S.L. and Hadgraft J. (2000) Int. J.
OSullivan, D. and Parmar, M.K. (2001) Thermochimica Acta, 380, Pharm., 198, 229-238.
13-17. [48] Lervolino, M., Cappello, B., Raghavan, S.L. and Hadgraft J. (2001)
[11] Irving, R.J. and Wads, I. (1964) Acta. Chem. Scand., 18, 195-201. Int. J. Pharm., 212, 131-141.
[12] Kilday, M.V. (1980) J. Res. Nat. Bur. Stand., 85, 467-481. [49] Barriocanal, L., Taylor, K.M.G. and Buckton, G. (2004) Int. J.
[13] Uriano, G.A. (1981) National Bureau of Standards Certificate. Pharm., 287, 113-121.
Standard Reference Material 1655, Potassium Chloride, KCl (cr) [50] Gaisford, S., Royall, P.G. and Greig, D.T.G. (2004)
for Solution Calorimetry. Thermochimica Acta, 417, 217-221.

You might also like