You are on page 1of 14

Subscriber access provided by AZ Library

Article
Solubility Advantage of Pharmaceutical Cocrystals
David J. Good, and Nai#r Rodri#guez-Hornedo
Cryst. Growth Des., Article ASAP • DOI: 10.1021/cg801039j • Publication Date (Web): 20 March 2009
Downloaded from http://pubs.acs.org on April 14, 2009

More About This Article

Additional resources and features associated with this article are available within the HTML version:

• Supporting Information
• Access to high resolution figures
• Links to articles and content related to this article
• Copyright permission to reproduce figures and/or text from this article

Crystal Growth and Design is published by the American Chemical Society. 1155
Sixteenth Street N.W., Washington, DC 20036
CRYSTAL
GROWTH
Solubility Advantage of Pharmaceutical Cocrystals & DESIGN
David J. Good and Naı́r Rodrı́guez-Hornedo* XXXX
Department of Pharmaceutical Sciences, College of Pharmacy, UniVersity of Michigan, VOL. XXX, NO.
Ann Arbor, Michigan 48109 XX
ReceiVed September 17, 2008; ReVised Manuscript ReceiVed December 22, 2008
000–000

ABSTRACT: Pharmaceutical cocrystals can improve solubility, dissolution, and bioavailability of poorly water soluble drugs.
However, true cocrystal solubility is not readily measured for highly soluble cocrystals because they can transform to the most
stable drug form in solution. The objectives of this study are to develop a method to estimate the cocrystal solubility in pure solvent
and establish the influence of constituent drug and ligand (i.e., coformer) properties. Cocrystal solubility and solubility product were
derived from transition concentration measurements where a solution is in equilibrium with solid drug and cocrystal. Transition
concentrations and solubilities are reported for carbamazepine cocrystals in water, ethanol, isopropanol, and ethyl acetate. The
aqueous solubility for seven carbamazepine cocrystals was estimated to be 2-152 times greater than the solubility of the stable
carbamazepine dihydrate form. Cocrystal solubility is shown to be directly proportional to the solubility of constituent reactants for
carbamazepine, caffeine, and theophylline cocrystals. The ligand transition concentration is also correlated with ligand solubility.
Transition concentration measurements reveal drug solubilization by ligand for several of the cocrystals studied. The correlation
established between constituent and cocrystal solubility was not effectively predicted by fusion properties of the various crystal
forms considered.

Introduction between cocrystal and solution that can be used to calculate


the often inaccessible equilibrium cocrystal solubility in pure
An important goal of solid-state pharmaceutical development solvent. The majority of studies relating to cocrystal solubility
is to increase drug solubility while maintaining a stable form. have focused on kinetic measurements of dissolution. Kinetic
This objective is critical because solubility and permeability are dissolution results are influenced by phase transformations,
the major factors used to describe oral absorption according surface area, and particle size distribution of the drug as well
to the biopharmaceutics classification system (BCS). Oral as fluid dynamics and experimental apparatus.10–13 These factors
absorption of BCS class II drugs is solubility-limited. This class can be difficult to quantify and reproduce. Kinetic and equi-
of drugs is currently estimated to account for about 30% of librium solubility measurements provide alternative and comple-
both commercial and developmental drugs.1,2 Class I and III mentary characterization of drugs useful for addressing oral
BCS drugs have high solubility and oral absorption primarily absorption limitations highlighted in the BCS. This paper focuses
limited by dissolution rate and permeability, respectively. on methods for equilibrium measurements of cocrystal solubility
Increased solubility can significantly improve the oral absorption that are experimentally accessible and reproducible.
of class II drugs and would have lesser impact for the already Earlier reports from our laboratory have shown that cocrystal
soluble class I and III drugs.3 Cocrystals have emerged as a solubility is dependent on the concentration of ligand in solution.
promising means to modify solubility, dissolution, and other The dissociation of cocrystal in solution is described by the
physicochemical properties of drug substances.4–8 Pharmaceuti- solubility product (Ksp), which is defined as a product of drug
cal cocrystals provide an alternative to chemical modification and ligand solution concentrations.14 This is analogous to the
of the drug substance as well as established salt, amorphous, Ksp of salt forms defined by the product of ionized drug and
solvate, and polymorphic drug forms that all have limitations counterion concentrations.13,15 Ksp is a constant that reflects the
in their utility. Cocrystals can be made for nonionizable drugs, strength of cocrystal solid-state interactions of drug and ligand
which are restricted from salt formation. Also, for ionizable relative to interactions with the solvent. The cocrystal solubility
drugs, the number of suitable cocrystal ligands (i.e., coformers) product behavior indicates that high ligand concentrations are
can exceed the number of suitable counterions. One example associated with low solution drug levels. In a similar manner
is the ionizable drug piroxicam, which has more than 50 reported salt forms of drugs exhibit a common-ion effect where solubility
cocrystal ligands.9 With unique properties for each drug form, decreases as the counterion concentration increases.13,16–18
there is great potential to form highly soluble and stable Cocrystal solubility is also a function of the solubility product
pharmaceutical cocrystals. such that high Ksp values equate to high cocrystal solubility.
The purpose of this work is to establish how cocrystal Exceptions to this solubility product behavior can occur when
solubility and stability are related to the properties of the pure the cocrystal does not dissociate to its components in solution,
components. To determine these relationships, methods are there is high solution complexation, or the drug is highly
developed to calculate the true equilibrium solubility of coc- solubilized by ligand.14,19,20
rystals, which are often not the most stable solid phase in The cocrystal transition concentration or eutectic point is a
solution. Indeed some of the most relevant pharmaceutical key parameter that establishes the regions of thermodynamic
cocrystals are more soluble than the pure drug form and stability of cocrystal relative to its components.14,21–24 This is
therefore prone to transformation when exposed to solvent. This an isothermal invariant point where two solid phases coexist in
paper addresses the need for a readily measurable equilibrium equilibrium with solution. The pharmaceutically relevant transi-
tion concentrations in this work involve the equilibrium of the
* Phone: (734) 763-0101. Fax: (734) 615-6162. E-mail: nrh@umich.edu. drug and cocrystal solid phases because the drug is often the
10.1021/cg801039j CCC: $40.75 © XXXX American Chemical Society
B Crystal Growth & Design, Vol. xxx, No. xx, XXXX Good and Rodrı́guez-Hornedo

(3) is to determine if crystal lattice properties such as melting


temperature and enthalpy are sufficient indicators of cocrystal
solubility.

Theoretical Calculations
Cocrystal Solubility (SCC) and Solubility Product (Ksp). For
cocrystal ARBβ where A is drug and B is ligand, solubility is described
by the chemical equilibrium of solid cocrystal with solution and the
corresponding equilibrium constant given by
K
ARBβ y\z RA(soln) + βB(soln)

R β
aA(soln)aB(soln)
K) (1)
aARBβ(s)
Figure 1. Diagram of carbamazepine-succinic acid cocrystal structure
with hydrogen bonds indicated by dashed lines. Each carboxylic acid Defining solid cocrystal activity as unity (aARBβ(s) ) 1) and assuming
of the succinic acid forms two hydrogen bonds with the carbamazepine the activity coefficients (γ) of A and B equal unity for low solute levels,
amide.28 eq 1 reduces to

Ksp ) [A]R[B]β (2)


least-water-soluble component (e.g., class II BCS drugs). It was
previously shown that ligand in excess of the molar ratio in the where Ksp is the solubility product of the cocrystal. Considering the
equilibrium reaction above, the mass balance for [A] ) RSARBβand [B]
cocrystal is needed to reduce the cocrystal solubility to equal ) βSARBβ can be substituted into eq 2 to provide the cocrystal solubility:
that of the drug.14 It was also predicted that cocrystals with
√K /R β
R+β
high Ksp values require high ligand concentrations to achieve SARBβ ) sp
R β (3)
equivalent drug and cocrystal solubilities.14 These concepts are
developed in the current manuscript to provide methods for Cocrystal solubility always refers to intrinsic solubility in pure solvent,
unless otherwise noted, as defined by eq 3. This derivation of intrinsic
calculating cocrystal solubility in pure solvent and understanding cocrystal solubility is based on chemical equilibria for all solvents in
relationships between transition concentrations, cocrystal, and which the drug and ligand substances are nonionizing. Extensive
component solubilities. considerations of ionization have been recently published and indicate
cocrystal Ksp is based on the concentrations of nonionized drug and
Carbamazepine (CBZ) cocrystals serve as the model system ligand; however, the expression of cocrystal solubility includes ap-
for this research. Carbamazepine is a widely used antiepileptic propriate pH and pKa factors for ionizing substances.29–33 One case is
drug that has low aqueous solubility and requires high blood the cocrystal of a neutral drug (e.g., carbamazepine) and an acidic ligand
for which solubility is given by
levels for therapeutic efficacy. The low bioavailability of

√ ( )
carbamazepine is due to low solubility and autoinduced Ksp Ka
R+β
metabolism.5,25 Carbamazepine has four known polymorphs and SARBβ ) R β
1+ (4)
a dihydrate form.26,27 More than 40 cocrystals of carbamazepine R β [H+]
have been identified to date.23,28 One recent publication where pH and ligand pKa are reflected in the hydrogen ion concentration
synthesized more than 25 carbamazepine cocrystals by screening and the acid dissociation constant.
Cocrystal Solubility (SCC) and the Phase Solubility Diagram
carboxylic acid ligands.23 Cocrystals of carbamazepine typically (PSD). Phase solubility diagrams and triangular phase diagrams are
form through hydrogen bonding of the primary amide group used to represent the solubility and stability of cocrystals. The PSD in
with a coformer. One example of this bonding is shown in Figure 2 represents two different cocrystals, which are either stable
Figure 1 for carbamazepine-succinic acid cocrystal. Pharma- (case 1, low solubility and Ksp) or metastable (case 2, high solubility
and Ksp) with respect to the pure drug form in a given solvent. These
cokinetic studies of carbamazepine cocrystallized with saccharin
curves represent cocrystal solubility product behavior with the drug
have shown blood level increases due to dissolution improve- concentration as a function of ligand given by [drug]R ) Ksp/[ligand]β
ment over the marketed pure drug (form III, monoclinic).5 The from eq 2. The drug solubility (horizontal line) is assumed to be much
carbamazepine cocrystals in this study have several ligands in lower than the ligand solubility, which is not shown. A dashed line
common with reported cocrystals of theophylline and caffeine. represents stoichiometric solution concentrations or stoichiometric
dissolution of cocrystal in pure solvent and its intersection with the
Both of these drugs are used to compare physicochemical cocrystal solubility curves (marked by circles) indicates the maximum
properties of carbamazepine cocrystals. Carbamazepine is a class drug concentration associated with the cocrystal solubilities. Unless
II BCS drug, whereas theophylline and caffeine are both class otherwise specified, the term cocrystal solubility refers to stoichiometric
III.2 solubility in pure solvent at conditions where the components are
nonionized. For a metastable cocrystal (case 2), the drug concentration
This paper has three principal objectives. Objective (1) is to associated with the cocrystal solubility is greater than the solubility of
enable the measurement of equilibrium cocrystal solubility from the stable drug form (horizontal line). The solubility of a metastable
cocrystal is not typically a measurable equilibrium and these cocrystals
a single measurement of solution in equilibrium with solid drug are referred to as incongruently saturating. As a metastable cocrystal
and cocrystal. This includes cocrystals that are either stable or dissolves, the drug released into solution can crystallize because of
metastable when exposed to pure solvent. Objective (2) is to supersaturation. This supersaturation is a necessary, but not sufficient
determine how the physicochemical properties of the reactants condition for crystallization. In certain instances, slow nucleation or
other kinetic factors might delay crystallization of the favored
are related to cocrystal solubility and stability. Included in this thermodynamic form and enable measurement of the true equilibrium
is understanding how diverse cocrystal solubility can be relative solubility. In the other case, a congruently saturating cocrystal (case
to alternative drug forms (e.g., salt or amorphous). Objective 1) has a lower drug concentration than the pure drug form at their
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX C

a variety of pharmaceutical analyses, there is a need for direct


information regarding the magnitude of thermodynamic cocrystal
solubility. For cocrystal-solvent systems where solubility, dissolution
behavior, or phase stability are unknown, determining the Ctr provides
the cocrystal solubility and solution stability. Additionally, Ctr values
can be measured with small amounts of drug because only a slight
excess of the solubility is needed to generate the requisite solid phases
in amounts detectable by a variety of analytical methods.
The solubility product expresses all possible solution concentrations
of drug and ligand in equilibrium with solid cocrystal and is directly
related to cocrystal solubility by eq 3. Inserting the cocrystal transition
concentrations ([A]tr and [B]tr) into eq 2 allows eq 3 to be rewritten as

√[A] [B] /R β
R+β R β
SARBβ ) tr tr
R β (5)
Equation 5 can be used to calculate cocrystal solubility from measure-
ment of the transition concentration. Drug and ligand transition
concentrations describe both the solubility and thermodynamic stability
of the cocrystal in a given solvent, and thus the Ctr is a logical first
Figure 2. Schematic phase solubility diagram of two different cocrystals
measurement for evaluating and comparing cocrystals. For the case
based on the Ksp of a stable (case 1) or metastable (case 2) cocrystal.
where drug solubility (SA) is unchanged with ligand concentration in a
Drug solubility is indicated and is much lower than the solubility of
particular solvent system, as shown in Figure 2, eq 5 can further simplify
the ligand, which is not shown. X marks represent the transition
by substitution of [A]tr with SA to give
concentrations (i.e., invariant point) used to calculate equilibrium
solubility. Circles represent the solubility of cocrystal in pure solvent.
√S [B] /R β
R+β R β
Dashed line illustrates stoichiometric concentrations of cocrystal SARBβ ) A tr
R β (6)
components that dissolution could follow. This line represents a drug
to ligand ratio equal to the cocrystal stoichiometric ratio of the The estimation of cocrystal solubility based on the ligand transition
components. concentration ([B]tr) and drug solubility is possible from eq 6. Equation
5 uses the full measure of Ctr (both [A]tr and [B]tr) to calculate cocrystal
respective solubility values. Therefore, the solubility of congruently solubility; however, the constant drug solubility assumption of eq 6 is
saturating cocrystals can be readily measured from solid cocrystal valid for many solute-solvent systems and can simplify the requisite
dissolved and equilibrated with solution. experimental methodology.
For both congruently and incongruently saturating cocrystals an Cocrystal Solubility and Chemical Potential at the Transition
isobarothermal invariant point (i.e., eutectic point), indicated by X marks Concentration. Cocrystal solubility is a function of chemical potential
in Figure 2, is the point where both solid drug and cocrystal are in (μ). The chemical potential expression for the equilibrium of cocrystal
equilibrium with a solution containing drug and ligand. Together the with solution is
drug and ligand solution concentrations at the invariant point are referred
to as the transition concentration (Ctr).14,19,23,34–37 Ctr values in this ARBβ(s) h RA(soln) + βB(soln)
paper define the solution concentrations ([drug]tr and [ligand]tr) that μAsolid
RBβ
) R(μAsoln) + β(μBsoln)
separate regions where either the solid cocrystal or drug are thermo-
dynamically stable. Other invariant points and transition concentrations At the transition concentration, the solution is saturated with drug (A).
exist to describe the equilibria between solid cocrystal and ligand, The chemical potential of solid drug and drug in solution are equal
cocrystals of different stoichiometry, or cocrystal solvates. Systems because they are at equilibrium.
where the ligand is less soluble than drug could utilize the eutectic
associated with solid cocrystal and ligand to readily calculate cocrystal μAsoln ) μAsolid
solubility.38 For CBZ and the high solubility ligands in this study, the
equilibrium of the solid cocrystal and drug is most relevant for When considering only one drug substance, μAsolid remains constant and
calculating the solubility of the cocrystal form in pure solvent. substituting μAsolid ) C ) μAsoln gives
Gibbs’ condensed phase rule defines a system with three components
(solvent, drug, and ligand) and three phases (drug, cocrystal, and μAsolid
RBβ
) β(μBsoln) + C (7)
solution) as having only one degree of freedom. At constant temperature
the phase rule indicates zero degrees of freedom (invariant point) where Therefore, the cocrystal chemical potential is proportional to that of
the solution composition is fixed (transition concentration). At the the ligand solute. From eq 7, it can be stated that cocrystal solubility
transition concentration, the free energy of solid cocrystal and drug is proportional to ligand solubility.
are equal (i.e., ΔG ) 0). Below or above the Ctr, either the drug or the Thermal Analysis and Predictions of Cocrystal Solubility (SCC).
cocrystal is less soluble, the free energy difference is nonzero, and only Melting temperatures and enthalpies of pharmaceutical crystals have
one solid phase is stable. A solid phase of cocrystal is stable beyond found prevalent utility as indicators of ideal solubility. These are readily
the Ctr, whereas pure drug is stable below this concentration. measurable properties associated with the crystal lattice energy that
Any cocrystal transition concentration wherein [drug]tr equals or must be overcome for dissolution to occur. Among structurally similar
exceeds [ligand]tr by a factor equivalent to the cocrystals’ stoichiometric pharmaceutical crystalline drug substances, those with high melt
coefficient ratio (R/β) is known to be congruently saturating provided temperatures are generally recognized to possess lower solubility.39,40
the ligand is more soluble, whereas all others are incongruently Equation 8 is a fundamental and common thermodynamic model for
saturating. If we assume Figure 2 represents a 1:1 cocrystal, where the relation between solubility and melt properties.40 For an ideal
drug is the least-soluble component in pure solvent, the transition solution, the solute solubility, x (mole fraction), is a function of the
concentration for the low-solubility cocrystal is above the dashed line heat of fusion, melt temperature, and solution temperature.
representing stoichiometric concentrations. Therefore, [drug]tr > [ligan-
d]tr, and the cocrystal is congruently saturating. -ΔHm (Tm - T)
The transition concentration is a readily measurable equilibrium state
ideal
ln xsolute ) (8)
R (TmT)
that can be used to determine cocrystal solubility. Furthermore, for
incongruently saturating cocrystals, the Ctr is the nearest accessible solid- where ΔHm = ΔHideal s
solution equilibrium to the solubility and a good surrogate measurement The two main assumptions in the derivation of this ideal solubility
from which to calculate cocrystal solubility. Only kinetic measurements expression are that enthalpy of fusion is temperature-independent (i.e.,
can provide alternative quantitative solubilities to those based on the heat capacity is zero) and approximately equal to the ideal enthalpy of
solubility product. Although these kinetic measurements are useful for solution. More involved expressions have been derived to address these
D Crystal Growth & Design, Vol. xxx, No. xx, XXXX Good and Rodrı́guez-Hornedo

Figure 3. (a) Flowchart of method used to establish the invariant point and determine equilibrium solution transition concentrations of cocrystal
components. (b) Schematic PSD illustrating two pathways to reach the Ctr marked with an X. The solubility curve is generated from drug and
ligand concentrations that equal the cocrystal solubility product.

assumptions by including additional thermodynamic parameters such method, a presaturated drug solution, which lies to the left of the Ctr
as solute heat capacity. The eq 8 approximation of ideal solubility, (graphically), is combined with solid cocrystal and drug. When starting
often regarded as an upper solubility limit, provides a good comparison with saturated drug solution the driving force to reach equilibrium is
for experimental cocrystal solubilities.39 cocrystal dissolution and the concurrent increase of ligand solute. As
the cocrystal dissolves, the drug released into solution is likely to
crystallize in the presaturated drug solution. Cocrystal dissolution may
Materials and Methods not lead to drug crystallization in instances where the ligand solubilizes
Materials. Anhydrous monoclinic form III carbamazepine (CBZ(III)) the drug. In these instances, when drug solubilization by ligand occurs,
as well as anhydrous theophylline (THP) and caffeine (CAF) were the excess solid drug initially added with the cocrystal maintains drug
obtained from Sigma-Aldrich (99+% purity) and used as received. saturation. Together these methods confirm the Ctr is reached by
Carbamazepine dihydrate (CBZ(D)) was prepared by stirring CBZ(III) converging to the same equilibrium from two different initial states.
in water for at least 24 h. Hydrates of theophylline and caffeine were Ctr values can be expressed as the range established from the two
prepared in the same manner. The cocrystal ligands nicotinamide methods. Steps for both methods to reach Ctr are:
(NCT), malonic acid (MLN), glutaric acid (GTA), saccharin (SAC), Cocrystal Precipitation Method Step 1: Prepare ligand solution
anhydrous oxalic acid (OXA), succinic acid (SUC), and salicylic acid solvent
(SLC) were obtained from Sigma-Aldrich (99+% purity) and used as B(s) 98 B(soln)(slightly undersaturated)
received. All crystalline drugs and ligands were characterized by X-ray
powder diffraction (XRPD) and Raman spectroscopy before carrying
out experiments. No impurities in the form of additional peaks were Step 2: Add excess drug
resolved during HPLC analysis of solutions containing the drugs or
B(soln)
ligands in this study. Ethanol (EtOH), isopropyl alcohol (IPA), and
A(s)xs y\z A(soln) + ARBβ(s)
ethyl acetate (EtOAc) were obtained from Fisher Scientific and dried
using molecular sieves prior to use. All the cocrystals used for solubility
studies were precipitated from ligand solutions by adding solid drug Cocrystal Dissolution Method Step 1: Presaturate solution with drug
according to the reaction crystallization method (RCM).21,23 Cocrystals
of CBZ-NCT and CBZ-SAC throughout the paper refer to the form I solvent
polymorphs.41 CBZ-MLN is the form B polymorph, which is a hydrated A(s)xs y\z A(soln)(saturated)
crystal form.23
Determination of Transition Concentrations ([A]tr and [B]tr). The
measurements of cocrystal Ctr values were preformed by precipitating Step 2: Add excess cocrystal
cocrystal as a result of stirring excess solid drug in a ligand solution A(soln)
wherein cocrystal synthesis occurs through RCM.21,23 Ctr values were ARBβ(s)xs y\z B(soln) + A(s)
also obtained by dissolution of cocrystal in saturated drug solutions
containing excess solid drug. Each cocrystal Ctr was determined from
both supersaturated cocrystal conditions by RCM and from undersatu- High-Performance Liquid Chromatography (HPLC). Solution
ration by cocrystal dissolution. Samples were confirmed to have two concentrations of drug and ligand were analyzed by Waters HPLC
solid phases (drug and cocrystal) at equilibrium for at least 24 h before (Milford, MA) equipped with a UV/vis spectrometer detector. A C18
the solution was isolated from the solids and analyzed by HPLC. A Atlantis column (5 μm, 4.6 × 250 mm; Waters, Milford, MA) at
flowchart of the processes used to determine cocrystal transition ambient temperature was used to separate the drug and the ligand. A
concentrations is given in Figure 3a. An aliquot of the solid phase was gradient method with a water, methanol, and trifluoroacetic acid mobile
isolated from solution for X-ray analysis then solid reactant(s) were phase was used with a flow rate of 1 mL/min. Sample injection volume
added as required to reach mixed solid phase equilibrium. This process was 20 μL. Absorbance of the drug and ligand analytes was monitored
was repeated in 24 h iterations until a stable mixed solid phase was between 210-300nm. Empower software from Waters was used to
achieved. Samples were held between 23 and 25 °C for all transition collect and process the data. All concentrations are reported in molality
concentration measurements. (moles solute/kilogram solvent).
Figure 3b is a PSD illustrating theoretical pathways for the two X-ray Powder Diffraction (XRPD). XRPD was used to identify
methods used to establish cocrystal Ctr values. The method utilizing crystalline phases after slurrying samples to determine cocrystal
RCM starts to the right of the Ctr (graphically) with a near saturated transition concentrations and confirm the proper two solid phases (drug
ligand solution to which excess drug is added. As the drug dissolves and cocrystal) were in equilibrium with the solution. XRPD patterns
in ligand solution, the cocrystal becomes supersaturated (i.e., solution of solid phases were recorded with a Rigaku MiniFlex X-ray diffrac-
reactant concentrations increase above the cocrystal solubility curve) tometer (Danvers, MA) using Cu KR radiation (λ ) 1.5418 Å), a tube
and crystallizes from solution. Precipitation of cocrystal continues until voltage of 30 kV, and a tube current of 15 mA. The intensities were
ligand becomes the limiting reagent and the excess solid drug remains measured at 2θ values from 2° to 30° with a continuous scan rate of
in equilibrium with the cocrystal formed. In the cocrystal dissolution 2.5°/min. Samples, prior to and after slurry reactions, were analyzed
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX E

by XRPD. Results were compared to diffraction patterns reported in of drug substances. Pudipeddi et al. have surveyed polymorph
literature or calculated from crystal structures published in the solubility ratios for 55 drug substances, some with multiple
Cambridge Structural Database. THP-NCT is the only cocrystal in this forms, to provide 81 solubility ratios and all the values fell below
study whose XRPD pattern has not been published to the best of our
knowledge. This diffraction pattern is provided in the Supporting five with only one exception.44 The highest polymorph solubility
Information. ratio was 23.1 (Premafloxacin I/III) followed by 4.7 (Acemetacin
Thermal Analysis. Crystalline samples of 3-6 mg were analyzed III/I). The solubility ratio of CBZ III/I polymorphs in IPA was
by differential scanning calorimetry (DSC) using a TA instrument almost 1.4. In comparison, the CBZ-GTA cocrystal ratio in IPA
(Newark, DE) 2910 MDSC system equipped with a refrigerated cooling is more than four times the solubility of CBZ III. Hancock and
unit. DSC experiments were performed by heating the samples at a Parks published amorphous drug solubility ratios (amorphous/
rate of 10 K/min under a dry nitrogen atmosphere. Temperature and
enthalpy calibration of the instruments was achieved using a high purity
crystalline) of seven compounds in a variety of aqueous and
indium standard. Hermetic aluminum sample pans were used for all organic solvent systems.45 The maximum measured solubility
measurements. The mean result of three or more samples is reported enhancement of amorphous drug or excipients was 24 times
for each substance. (glucose in methanol 20 °C) the crystalline form. Two other
Cocrystal samples for DSC analysis comprised several large crystals pharmaceutical compounds (glibenclamide and polythiazide) had
grown by slow partial evaporation of solutions containing the reactants. solubility ratios greater than five. Although the cocrystals in
These crystals were isolated from solution, washed, and characterized
by Raman microscopy before being combined to yield an adequate
this study show a greater range of equilibrium solubility ratios
mass for DSC analysis. than many of these surveyed, amorphous, polymorph, or salt
forms, the benefits in terms of dissolution rates are not known.
Results and Discussion The rate in which any of these more soluble solid forms
transform in solution could be a determining factor in their
Cocrystal Solubility from Transition Concentrations utility.
Cocrystal Solubility, Transition Concentration, and Ligand
Table 1 lists the transition concentration values ([drug]tr and
Solubility Relationship. Examination of results in Table 1
[ligand]tr) for a series of cocrystals measured at the invariant
reveal regular patterns in the effects of solubility of components
point where two solid phases (drug and cocrystal) are in
on transition concentrations and cocrystal solubilities. Within
equilibrium with aqueous or organic solution. All cocrystal Ctr
the series of carbamazepine cocrystals, it is observed that
values were confirmed by XRPD analysis of the solid phase,
experimentally measured [ligand]tr increases with ligand solubil-
isolated from equilibrium with solution, as exemplified in Figure
ity and that [ligand]tr can be orders of magnitude above [drug]tr.
4 for CBZ-SUC. In Figure 4, each of the isolated solid phases
On the basis of Ksp behavior, [ligand]tr in excess of [drug]tr is
contains the equilibrium mixture of CBZ-SUC cocrystal and
associated with cocrystal solubility greater than drug solubility.
CBZ (form III in organic solvents or dihydrate in water),
The relationship between cocrystal [ligand]tr and ligand solubility
indicating that these solids are both stable in solution.
is evident for carbamazepine cocrystals in water (Figure 5a).
Cocrystal Ksp and intrinsic solubility values, that represent
This relationship is maintained for other cocrystals and other
nonionized drug and ligand in solution, were calculated from
solvents in Figure 5b. Higher [ligand]tr values are associated
transition concentrations according to eqs 2 and 5, respectively.
with higher Ksp as predicted by eq 5 and previous models.14
For aqueous samples, the pH was first used to calculate the
nonionized concentration of drug and ligand at Ctr. Cocrystal Cocrystal solubility ratios or drug concentrations associated
solubilities were multiplied by the stoichiometric coefficient to with cocrystal solubility are directly proportional to ligand
provide the associated drug concentration and normalized with solubility, as expected from the derived chemical potential
the relevant stable crystalline drug solubility to provide solubility expression (eq 7). This is shown in Figure 5b, where both
ratios ([drug]Scc/Sdrug). For 1:1 cocrystals, this ratio is the same cocrystal and ligand solubilities are normalized by drug solubil-
as the cocrystal solubility divided by the drug solubility. ity and a clear trend is observed in all solvents studied. For
Solubility ratios emphasize the magnitude of change in cocrystals of the same drug and different ligands, the pattern is
solubility achieved by various cocrystals and facilitate com- maintained, i.e., high cocrystal solubility ratios for high ratios
parisons between different solvents. The CBZ cocrystals studied of ligand to drug solubilities. For this small series of 1:1
have a range of aqueous solubility ratios from 2 to 152. In cocrystals, it appears that a ligand solubility about 10 times the
organic solvents, where ligand solubilities are closer to the drug solubility is needed for cocrystal solubilities to be greater
solubility of CBZ, cocrystal solubility ratios have a lower range than drug solubility. 2:1 cocrystals demonstrate the same
between 0.1 and 4.4. The aqueous intrinsic solubility of CBZ- correlation (data not graphed); however, the slope is slightly
SAC is 1.2 × 10-3 m and the solubility of CBZ form III is 1.6 lower as anticipated from eq 5. For each cocrystal, the ratio of
× 10-3 m. Because saccharin is acidic (pKa ) 1.6 at 25 °C), ligand and drug concentrations at the transition concentration
the CBZ-SAC aqueous solubility from eq 4 increases greatly is also proportional with the ligand to drug solubility ratio. A
in the small intestine (e.g., more than 150 times at pH 6), but more detailed analysis would require the pH dependence of Ctr
CBZ form III does not. This CBZ-SAC aqueous solubility is and solubility and is the subject of a future publication.
consistent with previous in vivo animal studies by Hickey et Cocrystal solubility behavior was further examined in each
al. that show the cocrystal has higher CBZ blood levels relative solvent as shown in Figure 6. Cocrystal solubility ratio as a
to the marketed CBZ form III that persist several hours after function of ligand solubility for each solvent confirms that high
dosing, although high variability was observed for the pharma- ligand solubility equates to high cocrystal solubility. A notable
cokinetic para- exception is CBZ-NCT (positive deviation) in water where high
meters.5,42 Dissolution studies by Nehm et al. also have shown CBZ aqueous solubility enhancement occurs in the presence of
higher dissolution rate for CBZ-SAC relative to CBZ form III NCT(aq). In ethyl acetate (least polar solvent, Figure 6c), the
in pH 7 solutions.30 Table 1 lists ligand solubilities and the drug solubility order for three of the four ligands differ from the other
solubilities used to calculate solubility ratios in various solvents. solvents. Yet, cocrystal solubility remains a function of ligand
The solubility ratios of the cocrystals in Table 1 exceed ranges solubility. Similarly, a proportional relationship has been
previously reported for salt, polymorph, and amorphous forms suggested between the aqueous solubility of pharmaceutical salts
F

Table 1. Cocrystal Transition Concentrations ([drug]tr and [ligand]tr), Component Solubilities, and Calculated Cocrystal Ksp Values, Solubilities, and Solubility Ratiosa
ligand ionization J
A:B C D I solubility
cocrystal cocrystal stoichiometry [ligand]tr (m) [drug]tr (m) E F G H cocrystal ratio [drug]Scc/
(drug-ligand) (drug-ligand) solvent pH (mean ( range/2) (mean ( range/2) Sligand (m) Sdrug (m)b % nonionizedc pKa Kspd solubility (m)e Sdrug
CBZ-MLNf 2:1g water 1.4 1.5 × 100 ( 2 × 10-2 7.5 × 10-4 ( 5 × 10-6 1.5 × 10+1 4.6 × 10-4 96 2.8 8.1 × 10-7 5.9 × 10-3 26
CBZ-GTA 1:1 water 2.0 8.9 × 10-1 ( 4 × 10-2 3.3 × 10-3 ( 3 × 10-4 1.0 × 10+1 4.6 × 10-4 100 4.4 2.9 × 10-3 5.4 × 10-2 117
CBZ-NCT 1:1 water 6.0 8.5 × 10-1 ( 4 × 10-2 5.8 × 10-3 ( 5 × 10-4 7.5 × 100 4.6 × 10-4 100 3.5 4.9 × 10-3 7.0 × 10-2 152
THP-NCT 1:1 water 5.8 8.1 × 10-1 ( 3 × 10-2 2.5 × 10-1 ( 3 × 10-2 7.5 × 100 3.1 × 10-2 100 3.5 2.0 × 10-1 4.5 × 10-1 15
CBZ-OXA 2:1g water 1.3 2.8 × 10-2 ( 2 × 10-3 8.0 × 10-4 ( 5 × 10-5 1.3 × 100 4.6 × 10-4 50 1.3 9.0 × 10-9 1.3 × 10-3 5.7
Crystal Growth & Design, Vol. xxx, No. xx, XXXX

CBZ-SUC 2:1 water 3.0 1.8 × 10-2 ( 3 × 10-4 6.6 × 10-4 ( 2 × 10-4 5.4 × 10-1 4.6 × 10-4 94 4.2 7.4 × 10-9 1.2 × 10-3 5.2
CBZ-SAC 1:1 water 2.1 8.6 × 10-3 ( 9 × 10-5 6.8 × 10-4 ( 4 × 10-6 1.8 × 10-2 4.6 × 10-4 24 1.6 1.4 × 10-6 1.2 × 10-3 2.6
CAF-SLC 1:1 water 2.6 8.4 × 10-3 ( 1 × 10-3 1.4 × 10-1 ( 5 × 10-4 1.4 × 10-2 1.1 × 10-1 72 3.0 8.5 × 10-4 2.9 × 10-2 0.3
THP-SLC 1:1 water 2.6 5.3 × 10-3 ( 8 × 10-4 4.3 × 10-2 ( 3 × 10-3 1.4 × 10-2 3.1 × 10-2 72 3.0 1.6 × 10-4 1.3 × 10-2 0.4
CBZ-SLC 1:1 water 2.9 2.3 × 10-3 ( 1 × 10-4 6.4 × 10-4 ( 1 × 10-5 1.4 × 10-2 4.6 × 10-4 56 3.0 8.2 × 10-7 9.1 × 10-4 2.0
CBZ-GTA 1:1 IPA 5.4 × 10-1 ( 2 × 10-2 9.1 × 10-2 ( 3 × 10-3 3.6 × 100 5.0 × 10-2 - - 4.9 × 10-2 2.2 × 10-1 4.4
THP-NCT 1:1 IPA 1.1 × 10-1 ( 2 × 10-2 2.1 × 10-2 ( 5 × 10-3 6.3 × 10-1 3.0 × 10-3 - - 2.3 × 10-3 4.8 × 10-2 16
CBZ-NCT 1:1 IPA 6.0 × 10-2 ( 4 × 10-3 6.1 × 10-2 ( 2 × 10-3 6.3 × 10-1 5.0 × 10-2 - - 3.7 × 10-3 6.1 × 10-2 1.2
CBZ-SAC 1:1 IPA 9.0 × 10-3 ( 1 × 10-3 4.3 × 10-2 ( 6 × 10-3 1.6 × 10-1 5.0 × 10-2 - - 3.9 × 10-4 2.0 × 10-2 0.4
CBZ-SUC 2:1 IPA 1.3 × 10-3 ( 6 × 10-4 4.6 × 10-2 ( 4 × 10-3 5.5 × 10-1 5.0 × 10-2 - - 2.8 × 10-6 8.8 × 10-3 0.4
CBZ-GTA 1:1 EtOH 5.3 × 10-1 ( 1 × 10-2 1.8 × 10-1 ( 4 × 10-3 2.8 × 100 1.4 × 10-1 - - 9.4 × 10-2 3.1 × 10-1 2.2
THP-NCT 1:1 EtOH 2.0 × 10-1 ( 2 × 10-2 2.9 × 10-2 ( 4 × 10-3 1.1 × 100 1.8 × 10-2 - - 5.8 × 10-3 7.6 × 10-2 4.2
CBZ-NCT 1:1 EtOH 1.5 × 10-1 ( 9 × 10-3 1.4 × 10-1 ( 1 × 10-2 1.1 × 100 1.4 × 10-1 - - 2.2 × 10-2 1.5 × 10-1 1.1
CBZ-SAC 1:1 EtOH 3.5 × 10-2 ( 3 × 10-3 1.2 × 10-1 ( 2 × 10-2 2.4 × 10-1 1.4 × 10-1 - - 4.2 × 10-3 6.5 × 10-2 0.5
CBZ-SUC 2:1 EtOH 1.8 × 10-2 ( 3 × 10-3 1.4 × 10-1 ( 2 × 10-2 8.1 × 10-1 1.4 × 10-1 - - 3.5 × 10-4 4.5 × 10-2 0.6
CBZ-GTA 1:1 EtOAc 9.0 × 10-2 ( 1 × 10-2 9.9 × 10-2 ( 4 × 10-3 1.0 × 100 4.9 × 10-2 - - 8.9 × 10-3 9.4 × 10-2 1.9
CBZ-SAC 1:1 EtOAc 4.7 × 10-2 ( 4 × 10-3 5.4 × 10-2 ( 2 × 10-3 1.8 × 10-1 4.9 × 10-2 - - 2.5 × 10-3 5.0 × 10-2 1.0
THP-NCT 1:1 EtOAc 2.0 × 10-2 ( 1 × 10-3 9.7 × 10-3 ( 4 × 10-4 1.1 × 10-1 6.2 × 10-3 - - 2.0 × 10-4 1.4 × 10-2 2.3
CBZ-NCT 1:1 EtOAc 1.3 × 10-2 ( 2 × 10-3 5.0 × 10-2 ( 3 × 10-3 1.1 × 10-1 4.9 × 10-2 - - 6.7 × 10-4 2.6 × 10-2 0.5
CBZ-SUC 2:1 EtOAc 8.5 × 10-4 ( 2 × 10-4 4.3 × 10-2 ( 5 × 10-3 4.6 × 10-2 4.9 × 10-2 - - 3.1 × 10-8 2.0 × 10-3 0.1
a
Table is sorted by solvent and descending [ligand]tr. At Ctr, cocrystal and hydrated or anhydrous drug exist in equilibrium with solution. b Solubility of hydrated forms are indicated for aqueous samples.
c
Calculated for the measured pH using referenced pKa values.43 d Ksp units are m2 and m3 for 1:1 and 2:1 cocrystals, respectively. e Calculated using eq 5. f Form B (hydrated cocrystal).23 g Disordered crystal
A+B
structure that does not provide definitive stoichiometry.28 Indicated values reflect HPLC measurement of dissolved cocrystals. Calculations: H ) (C(F/100))B(D)A, I ) √H/(AABB), J ) (A × I)/E.
Good and Rodrı́guez-Hornedo
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX G

highly soluble ligands the ability to solubilize CBZ is


distinctly different. [CBZ]tr values range from 2 to 13 times
SCBZ(D), and this makes measuring [CBZ]tr critical to evaluat-
ing cocrystal solubility. For these cocrystals, [ligand]tr values
(Table 1) follow the order of ligand solubility (MLN > GTA
> NCT), but the [CBZ]tr, indicative of drug solubilization
by ligand, has an inverse order (NCT > GTA > MLN). NCT
has been previously shown to solubilize hydrophobic drug
substances.51 For the low-solubility CBZ ligands (OXA, SUC,
SAC), little or no difference is exhibited between cocrystal
solubility calculated from eqs 5 and 6.
Cocrystal Stoichiometry and Solubility. Carbamazepine
cocrystals of succinic, oxalic, and malonic acid are the only
2:1 (drug:ligand) cocrystals in Table 1, and they exhibit a
negative deviation from the trend between cocrystal and ligand
solubility. Because these ligands (more soluble than CBZ)
account for a lesser mole fraction of the cocrystal, they have
reduced capacity to impart high solubility to the cocrystal. From
eq 3, it is expected that cocrystals with high mole fractions of
Figure 4. XRPD patterns of reference materials and solid phases
isolated from suspensions at the transition concentration: (a) CBZ(III),
the least-soluble component will have less solubility enhance-
(b) CBZ(D), and (c) CBZ-SUC followed by solid phases isolated from ment over pure drug. The solubility value of a 2:1 cocrystal
(d) water, (e) ethanol, (f) ethyl acetate, and (g) 2-propanol. does not equal the equilibrium drug concentration as in the case
of a 1:1 cocrystal. Cocrystal stoichiometry defines the relation-
and the hydrophilicity of their organic counterions. Counterions ship between solubility and the equilibrium drug concentration
of erythromycin with extended alkyl chains (i.e., more hydro- achieved.
phobic) decreased the aqueous salt solubility.46 Similar results Cocrystal and Drug Solubility. The solubility of cocrystal
have been shown for counterion studies of lincomycin salts.47 and drug exhibited a direct proportionality, as did the solubility
Studies of other drugs are consistent with this trend and have of cocrystal and ligand. Figure 8a shows the aqueous solubility
shown that counterions with more hydroxyl groups (i.e., more of CBZ, theophylline (THP), and caffeine (CAF) cocrystals with
hydrophilic) can provide higher salt solubility.48,49 Alternatively, salicylic acid (SLC) as a function of the drug solubility. These
analysis of several flurbiprofen salts has not found a link results indicate the cocrystal solubility trend follows drug
between solubility and counterion hydrophilicity, but suggests solubility (CAF > THP > CBZ) for salicylic acid cocrystals.
that crystal lattice interactions could become stronger in some It is worth noting that SLC is less water soluble than CAF and
instances where counterions are more polar.50 Whether lattice THP but more soluble than CBZ. Cocrystal solubility is higher
energies and/or component solubilities predict cocrystal or salt than drug hydrate solubility for CBZ-SLC and higher than SLC
solubilities depends on the interplay between solid-state and solubility for CAF-SLC, and thus these cocrystals are incongru-
solute-solvent interactions. ently saturating. THP-SLC is congruently saturating, with both
Cocrystal Ligands That Increase Drug Solubility. Some components more soluble than cocrystal.
ligands are capable of solubilizing the drug substance with which Nicotinamide cocrystals of CBZ and THP listed in Table 1
they form cocrystals. The direct measurement of drug concen- are plotted in Figure 8b to further illustrate the relationship
tration is critical for accurately expressing the Ctr and calculating between cocrystal and drug solubility. Each numerical data point
cocrystal solubility. One example is the CBZ-NCT cocrystal represents the drug solubility and in each solvent the more
where CBZ is highly solubilized by NCT(aq) (Figure 6d and soluble drug has the higher cocrystal solubility. THP aqueous
Table 1). The [CBZ]tr is more than 10-fold the solubility of
solubility is greater than CBZ and THP-NCT has the higher
CBZ(D). Other ligands have minimal effect on drug solubility,
aqueous solubility (far right points). For all organic solvents,
with [CBZ]tr slightly above CBZ solubility in pure solvent, but
CBZ is more soluble than THP, and accordingly, the CBZ-NCT
this effect is prominent for CBZ-NCT in water. The CBZ
cocrystals are more soluble. Figure 8b additionally demonstrates
solubilization by NCT(aq), indicated by [CBZ]tr > SCBZ(D), leads
the effect of ligand solubility on cocrystal solubility. All
to higher cocrystal solubility than expected by considering NCT
cocrystal solubilities increase with NCT except for CBZ-NCT
aqueous solubility alone.
in water (negative deviation). It is clear the CBZ solubility
A series of CBZ cocrystal solubilities in aqueous media were
decrease in water overshadows a smaller relative NCT solubility
calculated by two approaches to highlight the importance of
increase. Together, these NCT cocrystals of CBZ and THP
considering [CBZ]tr. The first approach is using eq 3, where
indicate the combined effect drug and ligand solubility have
Ksp )[CBZ]trR[B]trβ and the second assumes [CBZ]tr ) SCBZ(D) so
that Ksp ) SCBZ R
[B]trβ . Figure 7 plots aqueous CBZ cocrystal on cocrystal solubility. Cocrystal solubility is proportional to
solubility ratios as a function of ligand solubility. A direct trend the solubility of both drug and ligand components.
between ligand and cocrystal solubility ratio is evident in Figure Accuracy of Solubility Product Measurements. The CBZ-
7 when it is assumed that [CBZ]tr ) SCBZ(D) (open squares). A NCT cocrystal Ksp values based on a single measurement of
notable exception is the cocrystal hydrate CBZ-MLN (nega- the Ctr in this paper are very similar to those previously
tive deviation) identified to have a more soluble 2:1 determined by measuring cocrystal solubility for a variety of
anhydrous form (data not shown). Several cocrystals exhibit ligand concentrations.14 Ksp values (Table 1) for CBZ-NCT in
large increases in solubility when calculated using measured EtOH, IPA, and EtOAc are within the experimental error of
[CBZ]tr. This is evident for cocrystals of the three most those previously reported. Although these deviations are mini-
soluble ligands (NCT, GTA, and MLN). For this group of mal, other systems could exhibit less accuracy.
H Crystal Growth & Design, Vol. xxx, No. xx, XXXX Good and Rodrı́guez-Hornedo

Figure 5. (a) Relationship between [ligand]tr and ligand solubility for CBZ cocrystals in water. Log axes are shown to aid visualization of the
individual points due to the large range of values. The linear regression for the untransformed data in Table 1 is r2 ) 0.986. (b) Ratio of ligand to
drug solubility plotted against the cocrystal solubility ratio (filled circles) and the ratio of ligand to drug transition concentrations (open circles). All
aqueous samples are shown in red. Several cocrystals with the same ligands are labeled.

One factor that may contribute to the error is the proximity a good estimate of the level of complexation or solubilization
of the transition concentrations to the concentrations of drug and, as a result, the true solubility. Accuracy could decrease
and ligand at the equilibrium cocrystal solubility in pure solvent. for ligands that exhibit high drug solubilization, as reflected by
There is close Ctr proximity if the ratio of [drug]tr to [ligand]tr Sdrug,[drug]tr, and Ctr is far from stoichiometric solubility. For
is near the stoichiometric ratio of components in the cocrystal. these instances, it would be prudent to measure complexation
When the equilibrium solubility is far away from the measurable constants or activity coefficients to ensure accuracy of Ksp and
transition concentration it is necessary to make large extrapola- solubility calculations.
tions based on ideal Ksp behavior. In this case, the solute When designing screening methods for cocrystal solubility,
activities may no longer be replaced by concentrations. Ad- it is possible to evaluate Ksp by measuring the drug and ligand
ditionally, solution complexation of cocrystal components can concentrations at any point where the solution is in equilibrium
be concentration-dependent. Solubility products based on transi- with solid cocrystal. However, by measuring the Ctr it is also
tion concentrations do not account for this solution complexation possible to establish the range of cocrystal thermodynamic
or solubilization. Ctr values only reflect the level of solubilization stability in solution. In either case, the prior discussion of Ctr
for that particular solution composition. Therefore, solubility proximity, complexation, and activity apply to the calculations
calculations from the transition concentrations may not provide of cocrystal solubility. Most pharmaceutically relevant cocrystals
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX I

Figure 6. Solubility ratio of CBZ cocrystal to CBZ(D) as a function of the constituent ligand solubility (molal). The graphs represent (a) ethanol,
(b) 2-propanol, (c) ethyl acetate, and (d) water. The points in each graph represent the cocrystal CBZ-ligand by the corresponding ligand component
(b, GTA; +, NCT; 2, SUC; 9, SAC).

have solubility higher than the drug and are incongruently for 2:1 cocrystals). This method is analogous to ideal solubility
saturating. This means measurements based on Ctr will have values of drug salt forms, wherein the melting enthalpy is
the closest proximity and the most accuracy for a single point normalized per mole of constituent ions.52
measurement used to calculate Ksp. Solubility product calcula- The association between ligand and cocrystal melt temper-
tions based on eutectic (i.e., Ctr) points have been used ature is scattered for the CBZ cocrystals in Table 2. These five
successfully to identify the phase stability and solution chemistry cocrystals show the same relation as an expanded group of
of enatiomers.36 eighteen CBZ cocrystals (single endotherm nonhydrate forms)
Ideal Solubilities from Thermal Properties. Cocrystal recently reported.23 The linear regression for ligand and cocrystal
melting temperature and enthalpy were obtained from analysis melting in the two studies (r2 ) 0.67-0.72) reflects a limited
of DSC data shown in Figure 9. These cocrystals show unique qualitative relationship. This regression value is in agreement
fusion properties relative to their constituent reactants. Two with previous correlations of ligand and cocrystal melt temper-
cocrystals (CBZ-SAC and CBZ-OXA) have lower melt tem- atures for different drug substances.53,54 Because cocrystal lattice
peratures than their pure components. CBZ-SAC and CBZ-OXA energy and melt temperature are based in part on the lattice
melt temperatures are 177.5 and 164.7 °C, respectively, and arrangement and intermolecular interactions, it is reasonable that
for pure components SAC Tm ) 229.7 °C, OXA Tm ) 191.2 ligand and cocrystal melt temperatures do not exhibit a strict
°C, and CBZ Tm ) 191.1 °C. The other five cocrystal melt correlation.
temperatures are very close to or between those of their Experimental reactant and cocrystal aqueous solubilities also
reactants. To determine the ideal solubility of cocrystals from show a qualitative trend with melting temperature in plots a
eq 8 the melting enthalpy was first normalized by the cocrystal and b in Figure 10, respectively. The organic solvents appear
stoichiometry. The cocrystal enthalpy of melting listed in Table to have a better correlation than the aqueous solubility values.
2 is the measured value divided by the number of moles of This observation seems reasonable given the potential of drug
reactant per mole of cocrystal (i.e., 2 for 1:1 cocrystals and 3 or ligand polar functional moieties for specific interactions with
J Crystal Growth & Design, Vol. xxx, No. xx, XXXX Good and Rodrı́guez-Hornedo

Figure 7. Aqueous solubility ratio of CBZ cocrystals to CBZ(D) (i.e.,


[drug]Scc/Sdrug) plotted against ligand solubility. Data labels indicate the
ligand component of the cocrystal. Cocrystal solubility calculated from
eq 5 (b, [drug]tr measured) or from eq 6 (0, [drug]tr approximated by
drug solubility (Sdrug) in pure solvent). *, hydrated cocrystal; †, 2:1
cocrystal stoichiometry.

water and therefore deviations from ideal solubility behavior.


The solubility of cocrystals in ethanol and isopropanol (from
Table 1) are quite linear (linear regression: r2 > 0.95) as a
function of melt temperatures listed in Table 2. This correlation
is apparent in Figure 10b, where melt temperature and cocrystal
solubility (log axis) are plotted for the four solvents. However,
from this limited data set, it would be speculative to consider
the correlation of melt temperature and solubility superior for Figure 8. (a) Aqueous solubility of salicylic acid cocrystals (with CBZ,
either cocrystals or reactants. The relation of cocrystal solubili- THP, or CAF) plotted against the solubility of the hydrated drug. (b)
ties and melt temperatures (Table 2 and Figure 10b) is stronger Solubility of NCT cocrystals of CBZ and THP in water, EtOH, EtOAc,
than reported for aqueous kinetic solubility and melt temperature and IPA plotted against the respective NCT solubility. The numerical
for cocrystals comprising similar acidic ligands.54 This data data points represent dug solubility from Table 1 in mmolal.
seems comparable to studies of salt forms that indicate qualita-
tive trends between solubility and melt temperature.40,50,52,55,56 Ratios of the ideal cocrystal and ideal drug solubilities listed
Previous reports of ephedrine salts have shown qualitative in Table 2 were on a similar order of magnitude to the solution
relations between log solubility and melt temperature with measurements in Table 1. The measured solubility ratios of CBZ
approximately similar levels of scatter seen in this cocrystal cocrystal/CBZ(III) are plotted in Figure 10d against the ideal
study.52 solubility ratio. These ideal ratios have significant discrepancies
Experimental cocrystal solubilities are not well correlated with aqueous experimental ratios for CBZ-NCT and CBZ-GTA
with ideal values derived from the melting enthalpy and cocrystals where the ligand is known to solubilize the drug
temperature of the crystals (Figure 10c). The ratios of substance. Correlations for organic solvents are significantly
experimental aqueous solubility to ideal solubility are listed better than aqueous samples. The thermodynamic ideal solubili-
in Table 2 to emphasize these deviations. Both aqueous and ties seem to more adequately quantify the relative change
organic solvents included in Figure 10c seem to suggest that between cocrystal and drug (Figure 10d) than absolute solubility
crystal fusion properties alone are not sufficient for predicting values (Figure 10c).
cocrystal solubility. The solution chemistry of cocrystals Solubility predictions have been reported for many poly-
appears to be critical for describing solubility behavior. The morph, amorphous, and salt forms. These predictions are often
reactants in Table 2 have a better correlation between based on the free-energy difference between the two solid forms.
experimental and ideal (linear regression r2 ) 0.87-0.92) This difference is typically estimated from fusion data combined
solubilities. Still, several instances of large deviations suggest in some cases with heat-capacity values. This type of solubility
ideal solubilities are not adequate indicators of drug and prediction for amorphous drugs mostly overestimates the
ligand solubility from solution measurements. The results measured solubilities and is complicated by transformations to
further illustrate that melting properties associated with the less-soluble drug forms. In the case of amorphous glibenclamide,
breaking of the crystal lattice are not sufficiently predictive the measured solubility ratio was 14 and the predicted ratio was
of cocrystal solubility relations that involve solvent interactions. 112-1652 times the crystalline solubility.45 Most measured
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX K

Figure 9. DSC for cocrystals of (a) THP-NCT, (b) CBZ-NCT (c) CBZ-GTA, (d) CBZ-SAC, (e) CBZ-SUC, (f) CBZ-SLC, and (g) CBZ-
OXA.

Table 2. Melt Temperature and Enthalpy Used in Calculation of Ideal Solubility and Comparisons with Experimental Solubility Values
exp aqueous ideal aqueous solubility ideal cocrystal
solubility (m)a solubility (m)b Tm (°C) ΔHm (kJ/mol) c
ratio (exp/ideal) solubility ratio (cocrystal/drug)
Reactants
glutaric acid (GTA) 1.0 × 10+1 13.5 97.7 20.7 7.4 × 10-1
nicotinamide (NCT) 7.5 × 100 4.9 130.6 23.8 1.5 × 100
succinic acid (SUC) 5.4 × 10-1 0.6 188.1 32.4 9.0 × 10-1
theophylline (THP) 5.8 × 10-2 1.8 273.6 19.0 3.2 × 10-2
salicylic acid (SLC) 1.8 × 10-2 1.9 160.9 27.1 9.5 × 10-3
saccharin (SAC) 1.1 × 10-2 0.3 229.7 32.1 3.7 × 10-2
carbamazepine (CBZ) 1.6 × 10-3 1.4 192.1 25.6 1.1 × 10-3
Cocrystals
THP-NCT 4.5 × 10-1 8.9 175.0 14.6 5.1 × 10-2 5.1
CBZ-NCT 7.0 × 10-2 1.8 160.8 27.6 3.9 × 10-2 1.3
CBZ-GTA 5.4 × 10-2 5.7 125.9 23.2 9.5 × 10-3 4.1
CBZ-SAC 1.2 × 10-3 1.3 177.5 27.5 9.2 × 10-4 1.0
CBZ-SUC 1.2 × 10-3 0.5 188.9 33.3 2.4 × 10-3 0.4
CBZ-SLC 9.1 × 10-4 2.6 160.1 24.6 3.5 × 10-4 1.9
CBZ-OXA 1.3 × 10-3 4.6 164.7 20.0 2.8 × 10-4 3.3
a
Measured solubility for anhydrous reactants and cocrystals (from Table 1). b Ideal solubility calculated from eq 8 using melt temperature and heat of
fusion. Mole fractions were converted to molality units in water. c The enthalpy of melting for cocrystals is normalized by moles of component
molecules (drug + ligand) per mole of cocrystal. All aqueous solubility, thermal data, and ratios listed are for anhydrous crystal forms.

amorphous solubility ratios were below the predicted ranges for cocrystals because they can exhibit solution complexation
that span about 1 order of magnitude (e.g., 48–455 for or solubilization of drug by other crystal components. Neither
polythiazide). Polymorphs have shown much closer agreement of these is applicable to polymorphs.
between predicted and actual solubility ratios than these
amorphous forms. This is possibly because of minimal free
Conclusions
energy differences between polymorphs, as opposed to large
differences for crystalline and amorphous forms. Polymorph This work developed methods to predict cocrystal solubilities
studies of ten drug substances by Pudipeddi and Serajuddin in pure solvent from measurement of transition concentrations
provided calculated solubility ratios in good agreement with (Ctr). These predictions are based on solubility product equations
experimental ratios.44 From our limited set, it appears ideal that include measured solution concentrations at Ctr, where solid
cocrystal solubility ratio predictions (Table 2 far right column) cocrystal and drug are in equilibrium with solution. Results show
show better agreement with solution-based solubility ratios that: (1) cocrystal solubility increases with the solubility of the
(Table 1) than is the case for amorphous systems. Cocrystals cocrystal components, (2) ligand transition concentrations
in this study also have a weaker correlation than previously cited increase with cocrystal and ligand solubilities for cocrystals of
polymorph studies. The potential for deviations could be high the same drug, and (3) ligand solubility about 10-fold higher
L Crystal Growth & Design, Vol. xxx, No. xx, XXXX Good and Rodrı́guez-Hornedo

Figure 10. Equilibrium cocrystal and reactant solubilities or solubility ratios in water (O), EtOH (0), IPA (X), and EtOAc (4). Solubility as a
function of melt temperature for (a) reactants and (b) cocrystals. (c) Experimental cocrystal solubility versus ideal solubility. (d) Experimental
against ideal cocrystal solubility ratio (cocrystal/CBZ form III).

than drug leads to cocrystal being more soluble than drug. sharing findings relating to several carbamazepine cocrystals
Although it is generally expected that solubility and melting used in this research.
point are correlated, the small series of cocrystals studied show
Supporting Information Available: X-ray powder diffraction
that solvent-solute interactions dominate over lattice energies, pattern of the THP-NCT cocrystal (PDF). This material is available
particularly in water. free of charge via the Internet at http://pubs.acs.org.
Transition concentrations are essential indicators of cocrystal
stability and solubility and provide useful insights for cocrystal Note Added after ASAP Publication. After this paper was
selection. Furthermore, pharmaceutical processes that involve published ASAP on March 20, 2009, a sentence was added to
solutions can be designed with an understanding of solubility the Results and Discussion section; the corrected version was
reposted on April 3, 2009.
and stability provided by Ctr values. Solution processes such as
cocrystal screening, synthesis, manufacture, formulation, and
dosing of cocrystal drug products are influenced by transition References
concentrations. The analysis of solution chemistry and phase
(1) Lipinski, C. A., Solubility in water and DMSO: issues and potential
behavior presented enables the calculation of true equilibrium
solutions. In Pharmaceutical Profiling in Drug DiscoVery for Lead
solubility and stability through a single measurement of solution Selection; Borchardt, R. T., Kerns, E. H., Lipinski, C. A., Thakker,
concentrations at Ctr under equilibrium conditions. D. R., Wang, B., Eds.; AAPS Press: Arlington, VA, 2004; pp 93-
125.
Acknowledgment. We acknowledge financial support from (2) Takagi, T.; Ramachandran, C.; Bermejo, M.; Yamashita, S.; Yu, L. X.;
the American Foundation for Pharmaceutical Education, Amidon, G. L. Mol. Pharm. 2006, 3 (6), 631–643.
Purdue-Michigan Consortium on Supramolecular Assemblies (3) Amidon, G. L.; Lennernas, H.; Shah, V. P.; Crison, J. R. Pharm. Res.
1995, 12 (3), 413–20.
and Solid State Properties, the Fred W. Lyons Jr. Fellowship,
(4) Bak, A.; Gore, A.; Yanez, E.; Stanton, M.; Tufekcic, S.; Syed, R.;
as well as the Warner Lambert/Parke Davis and Upjohn Akrami, A.; Rose, M.; Surapaneni, S.; Bostick, T.; King, A.;
Endowment Fellowships from The University of Michigan Neervannan, S.; Ostovic, D.; Koparkar, A. J. Pharm. Sci. 2008, 97
College of Pharmacy. We also acknowledge Scott Childs for (9), 3942–56.
Solubility Advantage of Pharmaceutical Cocrystals Crystal Growth & Design, Vol. xxx, No. xx, XXXX M

(5) Hickey, M. B.; Peterson, M. L.; Scoppettuolo, L. A.; Morrisette, S. L.; abstract W5205.
Vetter, A.; Guzman, H.; Remenar, J. F.; Zhang, Z.; Tawa, M. D.; (32) Rodrı́guez-Hornedo, N.; Nehm, S. J.; Jayasankar, A. Cocrystals:
Haley, S.; Zaworotko, M. J.; Almarsson, O. Eur. J. Pharm. Biopharm. Design, Properties and Formation Mechanisms. In Encyclopedia of
2007, 67 (1), 112–9. Pharmaceutical Technology, 3rd ed.; Informa HealthCare: London,
(6) McNamara, D. P.; Childs, S. L.; Giordano, J.; Iarriccio, A.; Cassidy, 2006; pp 615-635.
J.; Shet, M. S.; Mannion, R.; O’Donnell, E.; Park, A. Pharm. Res. (33) Cooke, C. L.; Davey, R. J. Cryst. Growth Des. 2008, 8 (10), 3483–
2006, 23 (8), 1888–97. 3485.
(7) Remenar, J. F.; Morissette, S. L.; Peterson, M. L.; Moulton, B.; (34) We adopt the term eutectic for ternary systems of two solid forms
MacPhee, J. M.; Guzman, H. R.; Almarsson, O. J. Am. Chem. Soc. and one liquid in equilibrium, consistent with the literature.35,36 This
2003, 125 (28), 8456–7. is not to be confused with original use of eutectic for describing binary
(8) Remenar, J. F.; Peterson, M. L.; Stephens, P. W.; Zhang, Z.; Zimenkov, phase behavior. The eutectic point for these ternary systems are
Y.; Hickey, M. B. Mol. Pharm. 2007, 4 (3), 386–400. alternartively referred to as an “iosbarothermal or isothermal invariant
(9) Childs, S. L.; Hardcastle, K. I. Cryst. Growth Des. 2007, 7 (7), 1291– point” (see ref 37) or our preferred terminology “transition concentra-
1304. tion”.
(10) Higuchi, W. I.; Parker, A. P.; Hamlin, W. E. J. Pharm. Sci. 1965, 54, (35) Jacques, J.; Wilen, S. H. Enantiomers, Racemates, and Resolution.;
8–11. Krieger Publishing Company: Malabar, FL, 1991.
(11) Serajuddin, A. T.; Jarowski, C. I. J. Pharm. Sci. 1985, 74 (2), 148– (36) Klussmann, M.; White, A. J. P.; Armstrong, A.; Blackmond, D. G.
54. Angew. Chem., Int. Ed. 2006, 45 (47), 7985–7989.
(12) Serajuddin, A. T.; Jarowski, C. I. J. Pharm. Sci. 1985, 74 (2), 142–7.
(37) Glasstone, S., Textbook of Physical Chemistry, 2nd ed.; MacMillan:
(13) Stahl, P. H.; Wermuth, C. G. International Union of Pure and Applied
London, 1948.
Chemistry Handbook of Pharmaceutical Salts: Properties, Selection,
And Use; Wiley-VCH: Weinheim, Germany, 2002; p xiv. (38) Reddy, L. S.; Bethune, S. J.; Kampf, J. W.; Rodriguez-Hornedo, N.
(14) Nehm, S. J.; Rodriguez-Spong, B.; Rodrı́guez-Hornedo, N. Cryst. Cryst. Growth Des. 2009, 378–385.
Growth Des. 2006, 6 (2), 592–600. (39) Grant, D. J. W.; Higuchi, T., Solubility BehaVior of Organic Com-
(15) Bhattachar, S. N.; Deschenes, L. A.; Wesley, J. A. Drug DiscoVery pounds; Wiley: New York, 1990; p liii.
Today 2006, 11 (21-22), 1012–8. (40) Yalkowsky, S. H. Solubility and Solubilization in Aqueous Media;
(16) Corrigan, O. I. Salt Forms: Pharmaceutical Aspects. In Encyclopedia American Chemical Society: Washington, D.C., 1999; 61 -77.
of Pharmaceutical Technology, 2nd ed.; Swarbrick, J., Boylan, J. C., (41) Porter III, W. W.; Elie, S. C.; Matzger, A. J. Cryst. Growth Des. 2008,
Eds.; Marcel Dekker: New York, 2002; p v. 8 (1), 14–16.
(17) Miyazaki, S.; Nakano, M.; Arita, T. Chem. Pharm. Bull. (Tokyo) 1975, (42) Shan, N.; Zaworotko, M. J. Drug DiscoVery Today 2008, 13 (9-10),
23 (6), 1197–204. 440–6.
(18) Miyazaki, S.; Oshiba, M.; Nadai, T. J. Pharm. Sci. 1981, 70 (6), 594– (43) MLN: Das, S. N.; Ives, D. J. G. Proc. Chem. Soc. London 1961, 373–
6. 374. GTA: Speakman, J. C. J. Chem. Soc. 1940, 855–859. OXA:
(19) Jayasankar, A.; Reddy, L. S.; Bethune, S. J. ; Rodrı́guez-Hornedo, N. Darken, L. S. J. Am. Chem. Soc. 1941, 63 (4), 1007–1011. SUC:
Cryst. Growth Des. 2009, 9 (2), 889-897. Pinching, G. D.; Bates, R. G. J. Res. Natl. Inst. Stand. Technol. 1950,
(20) Nicoli, S.; Bilzi, S.; Santi, P.; Caira, M. R.; Li, J.; Bettini, R. J. Pharm. 45 (6), 444–449. SAC: Newton, D. W.; Kluza, R. B. Drug Intell. Clin.
Sci. 2008, 97 (11), 4830-4839. Pharm. 1978, 12 (9), 546–554. SLC: Bray, L. G.; Dippy, J. F. J.;
(21) Rodrı́guez-Hornedo, N.; Nehm, S. J.; Seefeldt, K. F.; Pagan-Torres, Hughes, S. R. C.; Laxton, L. W. J. Chem. Soc. 1957, 2405–2408.
Y.; Falkiewicz, C. J. Mol. Pharm. 2006, 3 (3), 362–7. (44) Pudipeddi, M.; Serajuddin, A. T. J. Pharm. Sci. 2005, 94 (5), 929–
(22) Klussmann, M.; White, A. J. P.; Armstrong, A.; Blackmond, D. G. 939.
Angew. Chem., Int. Ed. 2006, 45 (47), 7985–7989. (45) Hancock, B. C.; Parks, M. Pharm. Res. 2000, 17 (4), 397–404.
(23) Childs, S. L.; Rodrı́guez-Hornedo, N.; Reddy, L. S.; Jayasankar, A.; (46) Jones, P. H.; Rowley, E.; Weiss, A. L.; Bishop, D. L.; Chun, A. H.
Maheshwari, C.; McCausland, L.; Shipplett, R.; Stahly, B. C. Cryst. J. Pharm. Sci. 1969, 58 (3), 337–9.
Eng. Comm. 2008, 10 (7), 856–864. (47) Magerlein, B. J. J. Pharm. Sci. 1965, 54 (7), 1065–7.
(24) Chiarella, R. A.; Davey, R. J.; Peterson, M. L. Cryst. Growth Des. (48) Agharkar, S.; Lindenbaum, S.; Higuchi, T. J. Pharm. Sci. 1976, 65
2007, 7 (7), 1223–1226. (5), 747–749.
(25) Kobayashi, Y.; Ito, S.; Itai, S.; Yamamoto, K. Int. J. Pharm. 2000, (49) Wells, J. I., Pharmaceutical Preformulation: The Physicochemical
193 (2), 137–46. Properties of Drug Substances; Horwood, E. , Ed.; Halsted Press:
(26) Lang, M.; Kampf, J. W.; Matzger, A. J. J. Pharm. Sci. 2002, 91 (4), Chichester, U.K., 1988; p 227.
1186–90. (50) Anderson, B. D.; Conradi, R. A. J. Pharm. Sci. 1985, 74 (8), 815–
(27) Rodriguez-Spong, B.; Price, C. P.; Jayasankar, A.; Matzger, A. J.; 820.
Rodrı́guez-Hornedo, N. AdV. Drug DeliVery ReV. 2004, 56 (3), 241– (51) Sanghvi, R.; Evans, D.; Yalkowsky, S. H. Int. J. Pharm. 2007, 336
74. (1), 35–41.
(28) Childs, S. L.; Wood, P. A. ; Rodrı́guez-Hornedo, N.; Reddy, L. S.; (52) Black, S. N.; Collier, E. A.; Davey, R. J.; Roberts, R. J. J. Pharm.
Bates, S.; Hardcastle, K. I. Cryst. Growth Des. 2008, accepted. Sci. 2007, 96 (5), 1053–1068.
(29) Bethune, S.; Huang, N.; Jayasankar, A.; Rodrı́guez-Hornedo, N. (53) Aakeroy, C. B.; Desper, J.; Scott, B. M. Chem. Commun. 2006, (13),
Understanding and Predicting the Effect of Cocrystal Components and 1445–7.
pH on Cocrystal Solubility. Cryst. Growth Des. 2009, submitted. (54) Stanton, M. K.; Bak, A. Cryst. Growth Des. 2008, 8, 3856–3862.
(30) Bethune, S. J. Thermodynamic and kinetic parameters that explain (55) Rubino, J. T. J. Pharm. Sci. 1989, 78 (6), 485–489.
solubility and crystallization of pharmaceutical cocrystals. Ph.D. (56) Thomas, E.; Rubino, J. Int. J. Pharm. 1996, 130 (2), 179–185.
Thesis, University of Michigan, Ann Arbor, MI, 2008.
(31) Nehm, S. J.; Jayasankar, A.; Rodrı́guez-Hornedo, N. AAPS J. 2006, CG801039J

You might also like