throbber
ELSEVIER
`
`Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`Advanced
`DRUG DELIVERY
`Reviews
`
`www .elsevier.com/locate/ drugdeliv
`
`Crystalline solids
`Sudha R. Vippagunta\ Harry G. Brittainb, David J.W. Granta·*
`
`Department of Pharmnceutics, College of Pharmncy, University of lvfinnesota, Weaver-Densford Hall, 308 Harvard Street S.E.,
`Minneapolis, MN 55455, USA.
`6Center for Phannaceutical Physics, 10 Charles Road, Milford, NJ 08848, USA
`
`Received 18 October 2000; accepted 21 December 2000
`
`Abstract
`
`Many drugs exist in the crystalline solid state due to reasons of stability and ease of handling during the various stages of
`drug development. Crystalline solids can exist in the form of polymorphs, solvates or hydrates. Phase transitions such as
`polymorph interconversion, desolvation of solvate, formation of hydrate and conversion of crystalline to amorphous fonn
`may occur during various phannaceutical processes, which may alter the dissolution rate and transport characteristics of the
`drug. Hence it is desirable to choose the most suitable and stable form of the drug in the initial stages of drug development.
`The current focus of research in the solid-state area is to understand the origins of polymorphism at the molecular level, and
`to predict and prepare the most stable polymorph of a drug. The recent advances in computational tools allow the prediction
`of possible polymorphs of the drug from its molecular structure. Sensitive analytical methods are being developed to
`understand the nature of polymorphism and to characterize the various crystalline forms of a drug in its dosage form. The
`aim of this review is to emphasize the recent advances made in the area of prediction and characterization of polymorphs and
`solvates, to address the current challenges faced by pharmaceutical scientists and to anticipate future developments.
`© 2001 Elsevier Science B.V. All rights reserved.
`
`Keywords: Crystallinity; Polymorphs; Hydrates; Solvates; Formulation; Drug substance; Phase transformation; Characterization
`
`Contents
`
`1. Introduction .. ........ ..... .... .... ..... .... ........ ..... ......... .... .... .... ......... ......... .... .... .... ......... ..... ... ......... ... . .... ..... .... .... ..... .... .... ..... .... .... ...
`2. Recent advances in the identification, prediction and characterization of polymorphs .. .. ...... ... ... . . .... .. .. .... .. ... ... . . ... ... .. ... ... . . ... ... .. ...
`2.1. Types of polymorphism....................................................................................................................................................
`2.2. Packing polymorphism .. . . . . .. . . . . . . . . . . . . .. .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. .. . . . . . . . . . . . . .. . . . . .. . . . . . . .. .. . . . . .. . . . . . . .. . . . . . . .. . . . . . . . . . . . . .. .. . . . . . . . . . . . . . .
`2.3. Conformational polymorphism..........................................................................................................................................
`2.4. Phase transformations in the solid state..............................................................................................................................
`2.5. Prediction of polymorphs ................................ ........ ...................... ................. .................................. ...................... ........ ...
`2.6. Directing the crystallization of specific polymorphs ......................................... ...................... ........ ........ .............................
`2.7. Characterization ofpolymorphs using a combination of analytical techniques.......................................................................
`3. Recent advances in the identification and characterization of hydrates and solvates ... ...................... ........ .....................................
`3.1. Introduction to solvates and hydrates.................................................................................................................................
`3.2. Structural aspects.............................................................................................................................................................
`
`4
`6
`6
`7
`8
`9
`11
`12
`13
`15
`15
`15
`
`*Corresponding author. Tel.: + 1-612-6243-956; fax: + 1-612-6250-609.
`E-mnil address: grantOOl@tc.umn.edu (D.J.W. Grant).
`
`0169-409X/0l/S - see front matter © 2001 Elsevier Science B.V. All rights reserved.
`PII: S0169-409X(0l)00097-7
`
`Argentum EX1027
`
`Page 1
`
`

`

`4
`
`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`3.3. Phase transformation of hydrates and solvates .. ........ ...................... ................. ............. ..................... .............................. ...
`3.4. Prediction of the formation of hydrates and solvates ...........................................................................................................
`3.5. Characterization of hydrates and solvates ...........................................................................................................................
`4. Current challenges and future directions....................................................................................................................................
`4.1. Origins of the challenges .. .... .. .. .... .. ... ... . . ... ... .. .... .. .. ...... ........ .... .... ... . . . .. ..... .... .. .. ...... ... ... . . .... .. . . .... .. .. .... .. ... ... . . ... ... .. .... .. .. ...
`4.2. Phase transformations during processing............................................................................................................................
`4.3. Degree of crystallinity .................................... ........ ...................... ................. .................................. ...................... ........ ...
`4.4. Characterization of mixtures of polymorphs .......................................................................................................................
`5. Conclusions............................................................................................................................................................................
`References . . . .. . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . .. .. . . . . . . . . . . . . .. . . . . . . . . . . . . .. .. . . . . .. . . . . . . .. . . . . .. .. . . . . .. .. . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. .. . . . . . . . . . . . . .. . . . . .. .. . . . . .
`
`17
`18
`18
`18
`18
`19
`21
`21
`23
`24
`
`1. Introduction
`
`Most orgamc and inorganic compounds of phar(cid:173)
`maceutical relevance can exist in one or more
`crystalline fonns. When applied to solids, the adjec(cid:173)
`tive, crystalline, implies an ideal crystal in which the
`termed unit cells, are repeated
`structural units,
`regularly and indefinitely in three dimensions in
`space. The unit cell has a definite orientation and
`shape defined by the translational vectors, a, b, and
`c, and hence has a definite volume, V, that contains
`the atoms and molecules necessary for generating the
`crystal. Each crystal can be classified as a member of
`one of seven possible crystal systems or crystal
`classes that are defined by the relationships between
`the individual dimensions, a, b, and c, of the unit cell
`and between the individual angles, a, /3, and y of the
`unit cell [1,2]. The structure of a given crystal may
`be assigned to one of the seven crystal systems, to
`one of the 14 Bravais lattices, and to one of the 230
`space groups [1]. All the 230 possible space groups,
`their symmetries, and the symmetries of their diffrac(cid:173)
`tion patterns are compiled in the International Tables
`for Crystallography [3].
`The common crystalline forms found for a given
`drug substance are polymorphs and solvates. Crys(cid:173)
`talline polymorphs have the same chemical com(cid:173)
`position but different internal crystal structures and,
`therefore, possess different physico-chemical prop(cid:173)
`erties. The different crystal structures in polymorphs
`arise when the drug substance crystallizes in differ(cid:173)
`ent crystal packing arrangements and/ or different
`conformations. The occurrence of polymorphism is
`quite common among organic molecules, and a large
`number of polymorphic drug compounds have been
`noted and catalogued [ 4-7].
`Solvates, also known as pseudopolymorphs, are
`
`crystalline solid adducts contammg solvent mole(cid:173)
`cules within the crystal structure, in either stoichio(cid:173)
`metric or nonstoichiometric proportions, giving rise
`to unique differences in the physical and pharma(cid:173)
`ceutical properties of the drug. If the incorporated
`solvent is water, a solvate is termed a hydrate.
`Adducts frequently crystallize more easily because
`two molecules often can pack together with less
`difficulty than single molecules. While no definite
`explanations can be given, possible reasons include
`adduct
`symmetry,
`adduct-induced confonnation
`changes, and the ability to form hydrogen bonds
`through the solvent molecules [2,8,9]. Desolvated
`solvates are produced when a solvate is desolvated
`and the crystal retains the structure of the solvate
`[10]. Desolvated solvates are less ordered than their
`crystalline counterparts and are difficult to character(cid:173)
`ize, because analytical studies indicate that they are
`unsolvated materials (or anhydrous crystal fonns)
`when, in fact, they have the structure of the solvated
`crystal forn1 from which they were derived [ 11].
`Because different crystalline polymorphs and sol(cid:173)
`vates differ in crystal packing, and/ or molecular
`conformation as well as in lattice energy and en(cid:173)
`tropy, there are usually significant differences in
`their physical properties, such as density, hardness,
`tabletability, refractive index, melting point, enthalpy
`of fusion, vapor pressure, solubility, dissolution rate,
`other thermodynamic and kinetic properties and even
`color [12]. Differences in physical properties of
`various solid forms have an important effect on the
`processing of drug substances into drug products
`[13], while differences in solubility may have impli(cid:173)
`cations on the absorption of the active drug from its
`dosage fonn [14], by affecting the dissolution rate
`and possibly the mass transport of the molecules.
`These concerns have led to an increased regulatory
`
`Page 2
`
`

`

`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`5
`
`interest in understanding the solid-state properties
`and behavior of drug substances. For approval of a
`new drug, the drug substance guideline of the US
`Food and Drug Administration (FDA) states that
`"appropriate" analytical procedures need to be used
`to detect polymorphs, hydrates and amorphous forms
`of the drug substance and also stresses the impor(cid:173)
`tance of controlling the crystal form of the drug
`substance during
`the various stages of product
`development [11]. It is very important to control the
`crystal form of the drug during the various stages of
`drug development, because any phase change due to
`polymorph interconversions, desolvation of solvates,
`formation of hydrates and change in the degree of
`crystallinity can alter the bioavailability of the drug.
`When going through a phase transition, a solid drug
`may undergo a change in its thermodynamic prop(cid:173)
`erties, with consequent changes in its dissolution and
`transport characteristics [ 15].
`Various phannaceutical processes during drug
`development significantly influence the final crys(cid:173)
`talline fonn of the drug in the dosage fonn. The
`various effects of pharmaceutical processing on drug
`polymorphs, solvates and phase transitions have been
`described in detail by Brittain and Fiese [ 16] and will
`be discussed in later chapters. Briefly, processes such
`as lyophilization and spray drying may lead to the
`formation of the amorphous form of drug, which
`tends to be less stable and more hygroscopic than the
`crystalline product. Also, processing stresses, such as
`drying, grinding, milling, wet granulation, oven
`drying and compaction, are reported to accelerate the
`phase transitions in phannaceutical solids. 1l1e de(cid:173)
`gree of polymorphic conversion will depend on the
`relative stability of the phases in question, and on the
`type and degree of mechanical processing applied.
`Keeping these factors in mind, it is desirable and
`usual to choose the most stable polymorphic form of
`the drug in the beginning and to control the crystal
`form and the distributions in size and shape of the
`drug crystals during the entire process of develop(cid:173)
`ment. The presence of a metastable form during
`processing or in the final dosage form often leads to
`instability of drug release as a result of phase
`transformation [ 17].
`Crystallization plays a critical role in controlling
`the crystalline form and the distribution in size and
`shape of the drug. The significance of crystallization
`
`mechanisms and kinetics in directing crystallization
`pathways of pharmaceutical solids and the factors
`affecting the formation of crystals have been re(cid:173)
`viewed in detail by vaiious researchers [12,18,19]. A
`crystalline phase is created as a consequence of
`molecular aggregation processes in solution that lead
`to the formation of nuclei, which achieve a certain
`size during the nucleation phase to enable growth
`into macroscopic crystals to take place during the
`growth phase. The factors affecting the rate and
`mechanisms by which crystals are formed are:
`solubility, supersaturation, rate at which supersatura(cid:173)
`tion and desupersaturation occur, diffusivity, tem(cid:173)
`perature, and the reactivity of surfaces
`towards
`nucleation. The various forces responsible for hold(cid:173)
`ing the organic crystalline solids together, such as
`nonbonded interactions and hydrogen bonding, have
`been discussed in detail by Byrn et al. [2] and Etter
`[20].
`Various analytical methods are being currently
`used to characterize the crystalline form of the drug
`during the vai·ious steps of processing and develop(cid:173)
`ment. 1l1ese methods have been reviewed recently in
`detail by many authors [7,10,21-25]. The single
`most valuable piece of information about the crys(cid:173)
`talline solid, including the existence of polymorphs
`and solvates, is the molecular and crystalline struc(cid:173)
`ture, which is determined by single-crystal X-ray
`diffractometry
`[2]. Powder X-ray diffractometry
`provides a "fingerprint" of the solid phase and may
`sometimes be used to determine crystal structure.
`Once the existence of polymorphism ( or solvate
`formation) is definitely established by single-crystal
`and powder X-ray diffractometry, spectral methods,
`such as Fourier transform infrared absorption (FTIR)
`spectroscopy, Fourier transfonn Raman scattering
`(FT Raman) spectroscopy, solid-state nuclear mag(cid:173)
`netic resonance (SSNMR) spectroscopy, ultraviolet
`and visible (UV-Vis) and/ or fluorescence spectros(cid:173)
`copy [23] may be employed for further characteriza(cid:173)
`tion. Of special significance are thermal methods,
`such as differential scanning calorimetry (DSC),
`thermogravimetric analysis (TGA) and optical micro(cid:173)
`scopy using a hot stage [24]. These methods are
`almost always employed for further characterization.
`Modulated
`(temperature)
`differential
`scanning
`calorimetry (MDSC) in combination with DSC and
`optical microscopy are able to identify the glass
`
`Page 3
`
`

`

`6
`
`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`transition of amorphous forms with much greater
`clarity and allow unique insights into the glass
`transitional and polymorphic behavior of drug sub(cid:173)
`stances [26].
`Because solid-state NMR spectroscopy can be
`used to study crystalline solids, as well as pharma(cid:173)
`ceutical dosage forms,
`this powerful method
`is
`finding increasing application in deducing the nature
`of polymorphic variations [27], such as variations in
`hydrogen bonding network and molecular confonna(cid:173)
`tions among polymorphs [28,29] and for the de(cid:173)
`tennination of molecular conformations and mobility
`of drugs in mixtures and dosage forms [2]. Solid(cid:173)
`state 13C-NMR in conjunction with the techniques,
`known as high power proton decoupling, cross
`polarization (CP), and magic-angle spinning (MAS)
`offers information not obtained readily by other
`13C-solid(cid:173)
`techniques. Recently,
`two-dimensional
`state NMR spectroscopy has been used to study the
`three conformational polymorphs of 5-methyl-2-[(2-
`nitrophenyl)amino]-3-thiophenecarbonitrile [30]. Use
`of two-dimensional NMR and total suppression of
`spinning side bands (TOSS) pulse sequences allowed
`the separation of isotropic and anisotropic chemical
`shifts for the three forms. This is a very powerful
`method for analyzing differences in the chemical
`environment and is finding increased application in
`the study of conformational polymorphism.
`With advances in analytical methods, the current
`focus of research in the solid-state area is to under(cid:173)
`stand polymorphism and pseudopolymorphism at the
`molecular level. Knowledge of the crystal packing
`arrangements and the various intermolecular forces
`involved in the different packing arrangements will
`help in the prediction and preparation of the most
`stable polymorphs of a given compound well in
`advance, to avoid surprises during product develop(cid:173)
`ment. A current emphasis is on the development of
`software to predict crystal structures of polymorphs
`from molecular structures. A thorough understanding
`of the physicochemical properties of polymorphs and
`solvates (hydrates) is of primary importance to the
`selection of a suitable crystalline form and develop(cid:173)
`ment of a successful pharmaceutical product. Bray et
`al. [31] have shown that, by thorough characteriza(cid:173)
`tion of four different crystalline forms of L-738,167,
`a fibrinogen receptor antagonist by various analytical
`
`techniques, it was possible to determine the suitabili(cid:173)
`ty of one or two forms for the development of
`pharmaceutical oral dosage forms.
`The present review aims to emphasize the recent
`advances made in the area of prediction and charac(cid:173)
`terization of polymorphs and solvates, attempts to
`address the current challenges and problems faced by
`pharmaceutical scientists and intends to anticipate
`future development. This review does not attempt to
`provide solutions to the problems but attempts to
`review comprehensively the advances made in recent
`years to help address these problems.
`
`2. Recent advances in the identification,
`prediction and characterization of polymorphs
`
`2.1. Types of polymorphism
`
`Based on differences in the thermodynamic prop(cid:173)
`erties, polymorphs are classified as either enantio(cid:173)
`tropes or monotropes, depending upon whether one
`form can transform reversibly to another or not. In
`an enantiotropic system, a reversible transition be(cid:173)
`tween polymorphs is possible at a definite transition
`temperature below the melting point. In a mono(cid:173)
`tropic system, no reversible transition is observed
`between the polymorphs below the melting point.
`Four useful rules have been developed by Burger and
`Ramburger [32,33]
`to determine qualitatively the
`enantiotropic or monotropic nature of the relation(cid:173)
`ship between polymorphs. These rules are the heat of
`transition rule, heat of fusion rule, infrared rule and
`density rule.
`If, by use of the above rules, it is established that
`the polymorphs of a particular drug are enantiotropic
`or monotropic, then the next goal is to define the
`thermodynamically stable (or metastable) domain of
`each crystalline phase of a substance as a function of
`temperature. The plot of the Gibbs free energy
`difference, !:::.G, against the absolute temperature, T,
`gives the most complete and quantitative information
`on the stability relationship of polymorphs [22], with
`the most stable polymorph having the lowest Gibbs
`free energy. The !:::.G between the polymorphs may
`be obtained using several techniques operating at
`
`Page 4
`
`

`

`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`7
`
`different temperatures, such as solubility [34] and
`intrinsic dissolution rate. Yu [35] has derived thermo(cid:173)
`dynamic equations to calculate /j,G between two
`polymorphs and its
`temperature slope from
`the
`melting data. This method is essentially an extension
`of the heat of fusion rule, which is based on
`statistical mechanics. Extrapolating /j,G to zero gives
`an estimate of the transition temperature, from which
`the existence of monotropy or enantiotropy is in(cid:173)
`ferred. The integration of different types of data
`provides the /j,G vs. T curve over a wide temperature
`range and allows the consistency between techniques
`to be checked [22]. Another approach to establish the
`order of stability among various polymorphs has
`been studied using pressure versus temperature plots,
`e.g., for sulfanilamide and piracetam [36]. This
`approach is based upon Ostwald's principle of least
`vapor pressure, according to which the stable poly(cid:173)
`morph exhibits
`the
`lowest vapor pressure. The
`accuracy of this approach to establish the stability
`hierarchy among the polymorphs has been shown to
`be very much dependent on the accuracy of the
`experimental data.
`In recent years, the main focus of research has
`been the characterization of polymorphs arising from
`structural differences in the crystal lattice. It has
`been established for some time that organic mole(cid:173)
`cules are capable of forming different crystal lattices
`through two different mechanisms. One of the mech(cid:173)
`anisms is termed packing polymorphism, and repre(cid:173)
`sents instances where conformationally relatively
`rigid molecules can be assembled into different
`three-dimensional structures through the invocation
`of different intermolecular mechanisms. The other
`mechanism is termed confonnational polymorphism
`and arises when a nonconfonnationally rigid mole(cid:173)
`cule can be folded into different arrangements, which
`subsequently can be packed into alternative crystal
`structures. The distinction between packing poly(cid:173)
`morphism and conformational polymorphism
`is
`somewhat artificial because different packing ar(cid:173)
`rangements impose different conformations on the
`molecules, however slight, and different conforma(cid:173)
`tions will inevitably pack differently. The structural
`aspects associated with polymorphs have been re(cid:173)
`viewed recently [2], as have the analogous features
`of solvate and hydrate systems [9]. In the next
`
`section, the results of some more recent investiga(cid:173)
`tions are discussed.
`
`2.2. Packing polymorphism
`
`An investigation into the structures and charge
`densities of two polymorphs of p-nitrophenol has
`been performed with the aim of deducing the differ(cid:173)
`ent modes of inter-molecular hydrogen bonding that
`lead to the formation of the two structures shown in
`Fig. la and b [37]. A detailed analysis of the charge
`density of the two forms indicates charge migration
`from
`the benzene ring region to
`the nitro and
`hydroxyl groups that accompanies the transfonnation
`of one form into the other. In addition, polarization
`of the oxygen lone-pair electrons was found to be
`substantially larger in the crystal forms than in the
`free molecule, resulting in considerably larger dipole
`moments in the solid state.
`During the study of a new crystal form (fonn I) of
`
`Fig. 1. Molecular packing diagrams of the (a) [3 polymorph of
`p-nitrophenol, (b) a polymorph of p-nitrophenol, showing 50%
`probability displacement ellipsoids ([37], reproduced with the
`permission of the American Chemical Society).
`
`Page 5
`
`

`

`8
`
`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`chlordiazepoxide, it was found that the heat of
`transition between the two forms (forms I and II) is
`rather modest, and kinetic factors permit the exist(cid:173)
`ence of the metastable phase [38]. Both structures
`contain four crystallographically independent mole(cid:173)
`cules linked in dimers through hydrogen bonding,
`but the dimers are packed differently to yield the two
`crystal forms. Because the dimers in the fundamental
`units are spaced differently in the two forms, it was
`proposed that the solid-state enantiotropic trans(cid:173)
`formation entailed rearrangement of the dimer units.
`A different approach has been taken during an
`evaluation of the different structures fanned by
`sulfathiazole [39]. Using a graph set approach to
`classify the known structural differences and simi(cid:173)
`larities among the various fonns, it became possible
`to identify packing motifs common to three of the
`four crystal structures. Fig. 2 shows the unit cells of
`the polymorphs I, II, III and IV, where molecules are
`paired as hydrogen-bonded dimers. At the end of the
`process, the authors were able to deduce possible
`links between the observed patterns of hydrogen
`bonding, processes of nucleation, and the crystal
`growth observed from a number of solvent systems.
`Interestingly, the analysis did not indicate a relation(cid:173)
`ship between the appearance of a particular poly(cid:173)
`morph from solution and the growth of its fastest
`
`growing surface. Rather, it appeared as if the differ(cid:173)
`ent solvents affected the process of polymorph
`formation through their effects on nucleation of the
`various forms.
`
`2.3. Conformational polymorphism
`
`The conformational polymorphism of the two
`forms of piroxicam pivalate has been studied in
`detail [40]. This compound is distinctive in that the
`high-melting form (polymorph 1) contains an un(cid:173)
`anticipated array of associated molecules bound as
`centrosymmetric dimers through hydrogen bonding,
`with the amido nitrogen atom acting as the donor and
`the pyridine nitrogen as the acceptor (Scheme 1,
`structure I). The low-melting form (polymorph 2)
`contains molecules of two distinct conformational
`states coexisting in the same crystal (Fig. 3), but
`linked through different hydrogen bonding arrange(cid:173)
`ments. This latter finding represents another unusual
`aspect of the crystallography of the substance.
`The inclusion of different solvent molecules in a
`crystal lattice can lead to the existence of different
`packing patterns, and has also been found to in(cid:173)
`fluence the molecular conforn1ation of paroxetine
`hydrochloride in two solvate forms [41]. One form
`
`N
`d_'I
`
`N02 H
`
`~~--r\
`s---{
`V
`
`C
`
`:
`
`0
`
`(a) Form I
`
`CH3
`
`II
`
`(b) Form II
`Fig. 2. r nit cells of four polymorphs (I, II, III and IV) of
`sulfathiazole showing hydrogen bonds, with the dimer structure
`clearly discernible ([39], reproduced with the permission of the
`Royal Society of Chemistry).
`
`Ill
`
`Scheme 1. Molecular structure of piroxican1 pi valate (I) [ 40],
`(II)
`5-methyl-2- [2-( nitropheny l )an1ino ]-3-thiophenecarbonitri le
`[30], 2' -acetamido-4' -[N,N-bis(2-methylcarbonylethyl)amino]-4-
`nitroazobenzene (III) [48].
`
`Page 6
`
`

`

`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`9
`
`hemihydrate phase. Crystals of the isopropanol sol(cid:173)
`vate decomposes in the open air at room tempera(cid:173)
`ture, because the isopropanol molecules are released
`easily through
`the channel. The hemihydrate is
`relatively stable.
`In an impressive fundamental study, the poly(cid:173)
`morphism of 5-methyl-2-[2-(nitrophenyl)amino]-3-
`thiophenecarbonitrile (Scheme l, structure II) has
`been catalogued [42,43] and discussed in detail [2].
`This compound was crystallized as six solvent-free
`polymorphs, each of which differed in the mode of
`packing and in molecular conformation. The differ(cid:173)
`ent confonners yielded sufficient perturbations on the
`respective molecular orbital so that a variety of
`crystal colors (red, orange, and yellow) were ob(cid:173)
`served. To obtain a more detailed evaluation of the
`relative stability, the authors considered a partition(cid:173)
`ing of polymorphic energy differences into lattice
`and conformational contributions, and were able to
`deduce general trends that appeared valid in the
`absence of hydrogen bonding. The act of crystalliza(cid:173)
`tion was found to feature an interplay of opposing
`forces, with perpendicular molecular conformations
`being favored in fluid solutions, while a preference
`for planar /high dipole conformers existed in most
`crystal forms, as shown in Fig. 4 [42]. The unusual
`polymorphism displayed by this system may result
`from one or more of the following factors:
`the
`preference for perpendicular conformations in solu(cid:173)
`tions, the preference for planar /high dipole confor(cid:173)
`mers in crystals, the formation of inter- and in(cid:173)
`tramolecular hydrogen bonds, and the thermody(cid:173)
`namic tendency towards low energy and high en(cid:173)
`tropy.
`
`2.4. Phase transformations in the solid state
`
`Studies of phase transfonnations in the solid state
`are important, because the sudden appearance or
`disappearance of a crystalline form can threaten
`process development, and can lead to serious phar(cid:173)
`maceutical consequences if the transformation occurs
`in the dosage forms. Hence, an understanding of the
`kinetics and mechanism of phase transformations is
`of practical importance. The rearrangement of mole(cid:173)
`cules into a new structure during phase transforma(cid:173)
`tion may or may not involve a solvent or vapor
`phase. To explain the mechanism of solid-solid
`
`Fig. 3. Conformations of the two independent molecules of
`piroxicam pi valate (I) in polymorph 2. Thermal ellipsoids are
`drawn at the 40% probability level. and H atoms are shown as
`spheres of arbitrary size ([40]. reproduced with the permission of
`the American Pharmaceutical Association).
`
`was obtained as a hemihydrate, and the other as the
`solvate of isopropanol (2-propanol). In the unit cell
`of the hemihydrate, one finds two protonated parox(cid:173)
`etine and two chloride ions together with one water
`molecule. Interestingly, the two paroxetine molecules
`are conformationally nonequivalent, and exhibit a
`number of different bond angles and torsion angles.
`In the other form, the unit cell contains one proton(cid:173)
`ated paroxetine molecule, one chloride ion, and one
`isopropanol molecule disordered along a molecular
`channel. Furthermore, the conformation of the parox -
`etine molecule in the isopropanol solvate is different
`from either molecular conformation observed in the
`
`Page 7
`
`

`

`10
`
`SR. Vippagunta et al. I Advanced Drug Delivery Reviews 48 (2001) 3-26
`
`Free
`molecules
`
`Cryatals
`
`More planar{
`
`high-dipole
`
`No intermolecular
`
`ON
`OP
`
`6
`
`4
`
`2
`
`0
`
`-2
`
`15
`.€ -,
`.:II!
`.;..
`0
`cS
`C
`UJ
`
`_..;.;.; __ ~ i . - - -~ ;Jm
`---
`
`Perpendicular YN
`low-dipole
`y
`
`Stabilized by inter-
`' - - - -Y
`molecular H bonds
`-4..._ __ _____________ __ _
`Free molecules Polymorphic crystals
`
`Fig. 4. Comparison of conformational energies and crystal ener(cid:173)
`the various polymorphs of 5-methyl-2-[2-(nitro(cid:173)
`gies of
`phenyl)arnino]-3-thiophenecarbonitrile (II). Form R (red prisms,
`m.p. 106.2°C). form Y (yellow prisms, m.p. 109.8°C), form OP
`(orange plates, m.p. 112.7°C), form ON (orange needles, m.p.
`114.8°C), form YN (yellow needles, m.p. not measurable) ([42],
`reproduced with the permission of the American Chemical Socie(cid:173)
`ty).
`
`physical transition, four steps have been proposed:
`(a) molecular loosening in the initial phase; (b)
`formation of an intermediate solid solution; (c)
`nucleation of the new solid phase and (d) growth of
`the new phase
`[2].
`In an
`interesting
`study,
`Skwierczynski [44] has proposed a two-environment
`model to describe the decomposition reaction kinet(cid:173)
`ics of a crystalline solid, aspartame. The decomposi(cid:173)
`tion reaction of aspartame is a simple unimolecular
`thermally-induced aminolysis and
`the
`reaction
`proceeds under anhydrous conditions, i.e., water is
`not a reactant [45]. This model links the chemistry of
`the solid-state reaction with the molecular mobility
`of the reactant as the reaction proceeds. The advan(cid:173)
`tage of this model is that it can be used to determine
`the shelf life of a product from kinetic data gathered
`at elevated temperatures. Apart from sol

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket