throbber
Advanced Drug Delivery Reviews 56 (2004) 275 – 300
`
`www.elsevier.com/locate/addr
`
`High-throughput crystallization: polymorphs, salts, co-crystals
`and solvates of pharmaceutical solids
`
`Sherry L. Morissette a,*, O¨ rn Almarsson a, Matthew L. Peterson a,
`Julius F. Remenar a, Michael J. Read a, Anthony V. Lemmo a, Steve Ellis a,
`Michael J. Cima b, Colin R. Gardner a
`
`a TransForm Pharmaceuticals, Inc., 29 Hartwell Avenue, Lexington, MA 02421, USA
`b Department of Materials Science and Engineering, Massachusetts Institute of Technology, Room 12-011, 77 Massachusetts Avenue,
`Cambridge, MA 02139, USA
`
`Received 21 August 2003; accepted 6 October 2003
`
`Abstract
`
`The concepts of high-throughput (HT) screening and combinatorial synthesis have been integrated into the pharmaceutical
`discovery process, but are not yet commonplace in the pharmaceutical development arena. Emerging strategies to speed
`pharmaceutical development and capture solid form diversity of pharmaceutical substances have resulted in the emergence of
`HT crystallization technologies. The primary type of diversity often refers to polymorphs, which are different crystal forms of
`the same chemical composition. However, diverse salt forms, co-crystals, hydrates and solvates are also amenable to study in
`HT crystallization systems. The impact of form diversity encompasses issues of stability and bioavailability, as well as
`development considerations such as process definition, formulation design, patent protection and regulatory control. This
`review highlights the opportunities and challenges of HT crystallization technologies as they apply to pharmaceutical research
`and development.
`D 2003 Elsevier B.V. All rights reserved.
`
`Keywords: High-throughput; Crystallization; Polymorph; Solvate; Salt; Co-crystal
`
`Contents
`
`1.
`2.
`3.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Development of high-throughput crystallization technologies .
`Applications of high-throughput crystallization screening in pharmaceutical research and development: case studies.
`3.1.
`High-throughput salt selection .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.2.
`Solid form discovery in highly polymorphic systems .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.
`Avoiding latent polymorphism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.4.
`Prediction of crystallization and polymorphism: applications to pharmaceutical form studies .
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`
`276
`278
`285
`285
`287
`290
`291
`
`* Corresponding author. Tel.: +1-781-674-7823; fax: +1-781-863-6519.
`E-mail address: morissette@transformpharma.com (S.L. Morissette).
`
`0169-409X/$ - see front matter D 2003 Elsevier B.V. All rights reserved.
`doi:10.1016/j.addr.2003.10.020
`
`Gilead 2015
`I-MAK v. Gilead
`IPR2018-00126
`
`

`

`276
`
`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`.
`.
`.
`.
`.
`Engineering of co-crystals
`3.5.
`Post-screening analyses and form selection .
`4.
`Summary and outlook .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`.
`.
`.
`.
`
`292
`295
`296
`297
`
`1. Introduction
`
`Active pharmaceutical ingredients (APIs) are fre-
`quently delivered to the patient in the solid-state as part
`of an approved dosage form (e.g., tablets, capsules,
`etc.). Solids provide a convenient, compact and gen-
`erally stable format to store an API or a drug product.
`Understanding and controlling the solid-state chemis-
`try of APIs, both as pure drug substances and in
`formulated products, is therefore an important aspect
`of the drug development process. APIs can exist in a
`variety of distinct solid forms, including polymorphs,
`solvates, hydrates, salts, co-crystals and amorphous
`solids. Each form displays unique physicochemical
`properties that can profoundly influence the bioavail-
`ability, manufacturability purification, stability and
`other performance characteristics of the drug [1].
`Hence, it is critical to understand the relationship
`between the particular solid form of a compound and
`its functional properties. Discovery and characteriza-
`tion of the diversity of solid forms of a drug substance
`provide options from which to select a form that
`exhibits the appropriate balance of critical properties
`for development into the drug product. Importantly,
`the desired properties may vary with each mode of
`delivery (i.e., oral, pulmonary, parenteral, transdermal,
`etc.), such that the solid form may differ for each
`optimized dosage form. Given these options, the
`choice and design of pharmaceutical solid forms can
`be critically important to successful drug development.
`Solid form discovery and design depends on the
`nature of the molecule of interest and type of physical
`property challenges faced in its development. The
`preferred solid form is generally the thermodynami-
`cally most stable crystalline form of the compound
`[1,2]. However, the stable crystal form of the parent
`compound may exhibit inadequate solubility or dis-
`solution rate resulting in poor oral absorption, partic-
`ularly for water-insoluble compounds. In this case,
`alternative solid forms may be investigated. For
`ionizable compounds, preparation of salt forms using
`pharmaceutically acceptable acids and bases is a
`common strategy to improve bioavailability [1,3,4].
`
`Like the parent compound, pharmaceutical salts may
`exist in several polymorphic, solvated and/or hydrated
`forms.
`Most APIs and their salts are purified and isolated
`by crystallization from an appropriate solvent during
`the final step in the synthetic process. A large number
`of factors can influence crystal nucleation and growth
`during this process, including the composition of the
`crystallization medium and the process(es) used to
`generate supersaturation and promote crystallization
`[1,5 – 13]. The most notable variables of composition
`and processing are summarized in Table 1. Solid form
`screening is used to understand the effects that these
`variables have on the polymorphic outcome of a
`crystallization experiment, so that a robust process
`can be identified to produce the desired crystal form.
`Traditionally,
`the study of solid form diversity of
`active compounds has relied on the use of a variety
`of common process methods for generation of new
`forms, coupled with modern characterization methods
`for analysis of the solids produced [2,14]. Most often,
`however, a combination of solvent recrystallization
`(cooling or evaporative, as well as slurry conversion)
`and thermal analysis (e.g., hot stage microscopy,
`differential scanning calorimetry) are employed for
`initial form screening. Such methods are inherently
`slow and only allow exploration of a small fraction of
`the composition and process space that can contribute
`to form diversity. Before suggesting a form for devel-
`opment, scientists may have carried out only a few
`dozen crystallization experiments and possibly pre-
`pared a handful of different salts of a compound. The
`main reasons for the limited number of experiments
`are the constraints on availability of compound and
`scientists’ analytical capacity in a given time frame,
`and they are therefore often forced to make form
`selection decisions on incomplete data. Accordingly,
`it
`is not surprising that unexpected and undesired
`outcomes can, and do, occur later on in development.
`Despite more than a century of research [15], the
`fundamental mechanisms and molecular properties
`that drive crystal form diversity, specifically the
`nucleation of polymorphic forms, are not well under-
`
`

`

`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`277
`
`Table 1
`Crystallization composition and processing variables [1,2,8]
`Process variablesa
`
`Composition type
`
`Thermal
`
`Anti-solvent
`
`Evaporation
`
`Slurry conversion
`
`Other variables
`
`n Heating rate
`
`n Anti-solvent
`type
`
`n Rate of
`evaporation
`
`n Solvent type
`
`n Mixing rate
`
`n Cooling rate
`
`n Maximum
`temperature
`
`n Rate of anti-
`solvent addition
`n Temperature
`of anti-solvent
`addition
`
`n Evaporation time
`
`n Carrier gas
`
`n Incubation
`temperature
`n Incubation
`time
`
`n Incubation
`temperature(s)
`n Incubation time
`
`n Time of anti-
`solvent addition
`
`n Surface-volume
`ratio
`
`n Thermal cycling
`and gradients
`
`n Impeller design
`
`n Crystallization
`vessel design
`(including
`capillaries, etc.)
`
`Polymorph/
`solvates
`
`n Solvent/
`solvent
`combinations
`n Degree of
`supersaturation
`n Additive type
`
`Salts/
`co-crystals
`
`n Counter-ion
`type
`
`n Acid/base
`ratio
`n Solvent/
`solvent
`combinations
`
`n Additive
`concentration
`
`n Degree of
`super-saturation
`n Additive type
`and concentration
`n pH
`n Ionic strength
`a Applicable to all types of screens.
`
`stood [13,16]. As a result, predictive methods of
`assessing polymorphic behavior of pharmaceutical
`compounds by ab initio calculations remain a formi-
`dable challenge. Even in cases where the existence of
`a crystalline form is predicted, the stability relative to
`other crystalline packing arrangements has been dif-
`ficult to estimate with accuracy [17]. Moreover, the
`prediction of packing structures for multicomponent
`(e.g., solvates, hydrates, co-crystals) or ionic systems
`is not yet possible [17]. Due to these limitations, solid
`form discovery remains an experimental exercise,
`where manual screening methods are employed to
`explore form diversity of a compound.
`the drug
`Control over solid form throughout
`development process is of paramount
`importance.
`Reliable preparation and preservation of the desired
`form of the drug substance must be demonstrated,
`and has become increasingly scrutinized by regula-
`tory agencies as more sensitive and quantitative
`solid-state analytical methods have become available
`[18]. Many strategies to influence and control the
`crystallization process to produce the solid form of
`interest have been reported. Some examples include
`stereochemical control using tailor-made auxiliaries
`[19 – 21], targeted solvent recrystallization [22 – 24],
`and templating using a variety of surfaces (e.g.,
`organic single crystal substrates [25], surfaces of
`metastable crystal faces [25,26],
`inorganic crystal
`
`surfaces [27] and polymeric materials [28]). Recent
`studies have also begun to uncover the role of
`reaction byproducts and other impurities in determin-
`ing polymorphic outcome and crystal properties
`[29 – 32], and in fact, it has been shown that in some
`cases such species can stabilize metastable crystal
`forms [33,34]. In addition, new processing methods
`continue to be developed to improve discovery and
`characterization of new forms, including precipitation
`by supercritical fluid [35,36],
`laser induced nucle-
`ation [37 – 39] and capillary crystallization [40 – 42].
`However, there remains a lack of fundamental un-
`derstanding of the nucleation process and the specific
`factors that contribute to crystallization of diverse
`forms of a compound [13,21,23]. In order to fully
`control the crystallization process, the link between
`the physical or chemical processes that
`influence
`nucleation and crystal growth needs to be better
`established. It is in this area that new experimental
`methodologies have the potential to enable develop-
`ment of this knowledge base.
`There is reason to believe that the already compli-
`cated landscape of pharmaceutical solid forms will
`become even more complex in the future. It is now
`increasingly appreciated that hydrogen bonded co-
`crystal structures between active agents and molecules
`other than water or solvent can be prepared. For
`example, co-crystals of aspirin, rac-ibuprofen and
`
`

`

`278
`
`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`rac-flurbiprofen have been prepared by disrupting the
`carboxylic acid dimers using 4,4V-bipyridine [43].
`These structures are formally molecular compounds
`(or co-crystals) but do not
`involve formation of
`covalent bonds or charge transfer from or to the active
`substance. Recent demonstrations of these principles
`with drug compounds have been published [43 – 45].
`Exploration of a given compound’s polymorphs,
`hydrates, solvates, salts, co-crystals and combinations
`of all of these appears intractable by conventional
`experimental methods, and as the number of potential
`methods for exploring and controlling crystal form
`diversity continue to expand, existing strategies will
`become increasingly inadequate. In an effort to un-
`derstand form diversity in a more comprehensive
`manner, high-throughput (HT) crystallization systems
`have recently been developed. This methodology uses
`a combinatorial approach to solid form generation,
`where large arrays of conditions and compositions are
`processed in parallel. Experiments are performed at
`small scale to reduce the material demand and to
`afford the largest number of conditions possible.
`The large number of crystallization trials performed
`in these experiments reflects the reality that nucleation
`rate has an extremely non-linear dependence on the
`experimental conditions, and as such, the probability
`of a chance occurrence of a particular form is in-
`creased by a HT approach. Supersaturation (solubility)
`and induction time of the various possible solid forms
`are independently controlled by these conditions,
`resulting in highly non-linear time dependence of
`crystallization. In addition,
`the combinatorial ap-
`proach permits exploration of a chemical continuum,
`where use of many solvent mixtures may allow one to
`assess what underlying physical or chemical processes
`are required to produce a particular solid form. Once a
`variety of conditions that can be used to produce a
`given crystal form on the microscale are identified in
`the HT screen, scale-up studies are typically con-
`ducted to optimize the process for laboratory scale
`production.
`In this review, the development and application of
`novel HT crystallization technologies for exploration
`of solid form diversity are discussed. The operational
`features of a fully integrated, automated HT crystal-
`lization system are presented, highlighting the design
`requirements for hardware and software components,
`as well as general specifications for consumables.
`
`Case studies are used to illustrate the benefits and
`capabilities of the approach, including salt selection in
`early lead optimization (ELO) and pre-clinical devel-
`opment, polymorph and solvate screening in highly
`polymorphic systems, comprehensive discovery of
`crystal forms to reduce the risk of late displays of
`polymorphism, comparison of experimental and pre-
`dictive methods of solid form discovery, and engi-
`neering of co-crystals. The need for post-screening
`characterization of crystal forms to enable ranking and
`selection of the most suitable form for development is
`briefly reviewed. Finally,
`the implications of HT
`crystallization technologies on the future of solid form
`screening processes, intellectual property protection
`and regulatory compliance are discussed.
`
`2. Development of high-throughput crystallization
`technologies
`
`HT crystallization systems have been developed to
`more rapidly and comprehensively explore the multi-
`parameter space that contributes to solid form diver-
`sity [40,46 – 51]. In its simplest description, HT
`crystallization can be broken down into three key
`experimental steps: design of experiment (DOE),
`execution of experimental protocols and analysis of
`data. Systems designed to carry out these experiments
`generally consist of both hardware and software
`components that drive and track experimentation,
`and permit data storage, retrieval and analysis. Such
`systems should be designed to be flexible and scalable
`to ensure that a variety of experimental procedures
`can be carried out either serially or concurrently.
`Thus, the system can be employed at various stages
`of drug development, where differences exist in the
`quality and quantity of compound available. While it
`is highly desirable to have the ability to mine and
`model experimental data, and to use the subsequent
`knowledge to guide further experiments, not all HT
`crystallization systems are equipped with these fea-
`tures. In Section 3, the hardware and software con-
`siderations for design and development of a fully
`integrated, informatics-driven HT crystallization sys-
`tem are described.
`While the concepts of HT screening are widely
`applied in the pharmaceutical industry, most notably
`in the drug discovery arena [52], the application of
`
`

`

`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`279
`
`in particular
`HT approaches to drug development,
`solid form screening, are just beginning to be real-
`ized. These latter approaches, however, are more akin
`to HT experimentation than HT screening. Hence,
`several important distinctions, which reflect on the
`design of HT experimental systems, need to be made.
`First,
`the goal of HT screening is to get a small
`number of successful outcomes, which are then
`passed on to the next stage of development. Little
`effort is typically made to learn why certain outcomes
`were positive and why others were negative. In
`contrast, HT experimentation, such as HT crystalliza-
`tion, is carried out with the goal of having each point
`in the experiment produce multiple types of data that
`can be interpreted, and the interpretation used to
`guide the experimental process to a successful con-
`clusion. Second, unlike traditional HT screening
`assays where experiments are generally conducted
`under constant experimental conditions, HT crystalli-
`zation experiments for solid form discovery are best
`conducted using a variety of process methods, each
`having varying experimental conditions (e.g., temper-
`ature variations as a function of time) over the course
`of the experiment. These additional process variables
`permit maximal diversity in the experimental space,
`increasing the likelihood that comprehensive cover-
`age will be achieved. Finally, there is a distinction to
`be made in terms of relative ‘‘hit rates’’. In both HT
`screening and HT crystallization, a ‘‘hit’’ can be
`
`thought of as a set of conditions that gives rise to a
`desired result. In HT screening, the desired result is
`typically an activity, or potency, that exceeds a pre-
`defined threshold. In HT crystallization, a hit
`is
`defined as the formation of a solid. The typical
`observed hit rate of HT screening is on the order of
`0.1% of the total number of samples analyzed. In
`contrast, HT crystallization experiments can yield hit
`rates ranging from tens of percents to nearly 100%,
`depending on the type of experiment and the process
`mode(s) used. For example, while only a handful of
`compounds from a selection of thousands may exhibit
`the required potency, 10 – 50% of crystallization trials
`may yield solids. In fact, the range of wells that yield
`solids is very wide, depending on process mode and
`experimental
`time scale, as will be discussed in
`subsequent sections. The impact of these differences
`is manifested in the design and operational require-
`ments of HT experimentation systems.
`A fully integrated HT crystallization system con-
`sists of a number of components, including experi-
`mental design and execution software, robotic
`dispensing and handling hardware, automated high-
`speed micro-analytical tools, end-to-end sample track-
`ing and integrated cheminformatics analysis software
`for data visualization, modeling and mining. A sche-
`matic overview detailing the workflow of such a
`system is depicted in Scheme 1 [53]. These features
`are supported by a comprehensive informatics foun-
`
`Scheme 1. A schematic illustration of the workflow of a fully integrated HT crystallization system [53].
`
`

`

`280
`
`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`dation that is used to handle the large quantities of
`data generated. Specifically, informatics tools are used
`to design statistically relevant and diverse experi-
`ments, drive the automation hardware to perform the
`specified operations, and provide an analytical func-
`tion to analyze, compare and sort
`the results of
`experiments. An important feature of these systems
`is the ability to mine and model experimental data and
`use the knowledge generated to guide further experi-
`ments. These functions are supported by use of a
`relational database that provides a mechanism of
`communication between system components.
`When designing a HT crystallization experiment, or
`set of experiments, a large variety of parameters of
`composition and process are involved. Experimental
`designs must be aimed at covering a large multifacto-
`rial parameter space, with the goal of determining
`which experimental factors affect the desired outcome.
`In practice, it is desirable to place constraints on the
`experimental space, making common statistical design
`methods such as full or partial factorial designs inap-
`propriate or impractical. For example, hardware limi-
`tations, including minimum and maximum dispense
`volumes or masses and accessible temperature ranges,
`as well as constraints related to chemical compatibility
`(i.e., reactivity of components, miscibility, etc.) or
`toxicity limits of components (if appropriate), need
`to be considered. Thus, alternative DOE methods that
`can accommodate such constraints are required. D-
`optimal design [54,55] is an example of a DOE
`algorithm that can take a set of constraints, such as
`the ones described above, in combination with a target
`analytical model and determine the optimal set of
`experimental points to test. Another commonly used
`DOE algorithm is diversity generation, with which the
`experimentalist selects a set of pertinent chemical
`properties and uses the algorithm to evenly spread
`experimental points over the chosen property space. In
`addition, some systems utilize a solubility calculator
`tool to estimate the solubility of the API in the given
`solvent/additive mixture. The calculated information is
`then used to select the appropriate concentration of
`API in each mixture so that it is supersaturated with
`respect to the reference phase at the harvest tempera-
`ture. Here, the driving force for crystallization can also
`be varied by tailoring the composition of each sample
`based on the API solubility in that mixture. With such
`DOE tools, experiments may be designed to effective-
`
`ly and simultaneously explore the diverse composition
`and process space described in Table 1.
`Ideally, DOE algorithms should also incorporate
`prior knowledge or experimental results, which have
`been stored in a database as a set of rules or models, to
`limit an experimental space to have certain predicted
`characteristics. For example, over the course of time, a
`regression model may be developed between a set of
`known or calculated chemical properties and a pa-
`rameter of experimental interest. The model could be
`used during the design of a new experiment in order to
`test only those chemicals that are predicted to give a
`desirable result. Since a large number of factors need
`to be considered during experimental design, the DOE
`interface available to the scientist must not only be
`flexible and easy to use, but must also offer tools that
`aid design efficiency and effectiveness and permit
`input of scientific knowledge generated over time.
`At the end of the experimental design process, the
`resulting set of experimental conditions is translated
`into a series of commands for the HT systems, and
`stored in a relational database for later retrieval by the
`software that controls the automation. When an ex-
`periment is activated,
`the overall operation of the
`automation systems is managed by the HT informatics
`system, which is responsible for physical operation of
`the HT platforms as well as data tracking and storage.
`Execution of experimental commands is carried out
`by automated laboratory equipment that comprises the
`HT crystallization system. Specialized automated sys-
`tems perform several of the functions in a sequence of
`events that make up the experiment. Each station is
`controlled through an interface to the informatics
`system that ensures the samples are processed at the
`correct stations, in the correct order, with the selected
`experimental parameters being followed. Parameters
`of operation are recorded, including the time at which
`an action is taken. After execution of the experimental
`steps, the software interface retrieves any pertinent
`information generated by the automated platform,
`such as assay results or operational parameters, stores
`these data in the relational database, and updates the
`status of the experiment to reflect the completion of
`operations.
`In general, the hardware required for a HT crystal-
`lization system is comprised of four major functional
`elements: sample preparation, solids generation, solids
`detection and sample analysis. Sample preparation
`
`

`

`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`281
`
`involves adding the compound of interest (API) to the
`diverse set of conditions used to conduct crystalliza-
`tion studies. Typically,
`the API is dispensed as a
`solution in a suitable solvent, followed by solvent
`removal to yield the solid API. Solvent removal can
`be achieved by passive evaporation or by controlled
`active evaporation (e.g., use of a vortex dryer).
`Alternatively, the API can be delivered in the solid
`state with suitable powder handling systems. Depend-
`ing on the amount of saturation desired, the crystal-
`lization vessel used, and the API’s solubility in
`solvents or solvent mixtures of interest, API masses
`ranging from a few hundreds of micrograms to several
`milligrams will be present in each vessel. Once the
`API has been delivered to the crystallization vessels
`(tubes, vials or microwell plates), combinations of
`solvents and/or additives are added to each vessel. By
`taking advantage of the power of combinatorial
`approaches, large numbers of unique combinations
`can be dispensed from manageable sets of starting
`materials.
`Compatibility of equipment components (syringes,
`dispense tips, tubing, etc.) and consumables (plates,
`tubes, etc.) with solvents and other compounds is a
`key hurdle faced in the development of combinatorial
`crystallization for small molecules. Unlike protein
`crystallization systems [56,57], which are commonly
`based on the sitting-drop method in aqueous media,
`small molecule crystallization employs a range of
`crystallization additives and processes. The additives
`include organic solvents with varying properties (e.g.,
`alcohols, acetone, hexane, ethyl acetate, etc.), water,
`acids, bases and co-crystal formers, as well as other
`compounds (e.g., small molecule templating agents,
`surfactants, pharmaceutical excipients, etc.). This
`wide range of materials needs to be handled by
`appropriate liquid handling techniques to enable the
`combinatorial assembly previously mentioned. Ideal-
`ly, liquid transfers are achieved using multichannel
`pipettors with individually controllable channels.
`Depending on the crystallization vessel design, the
`volumes of reagents dispensed will be as low as a few
`microliters to as high as several hundred microliters.
`Potential for cross-contamination and tendency
`toward unwanted solvent evaporation from crystalli-
`zation wells are challenges that need to be addressed
`in a HT crystallization system. A large number of the
`solvents used to crystallize small molecules have high
`
`vapor pressure under ordinary laboratory conditions.
`Sealing of the crystallization vessels is key to being
`able to control composition during crystallization
`from these solvents. Due to solvent fugacity, vessels
`need to be protected from ingress of the components
`of neighboring wells. These problems have been
`solved by different means, such as sealing of individ-
`ual tubes with a Teflon-backed crimp seal [40] or O-
`rings/gasket seals and clamped covers [47,51].
`HT crystallization must enable several process
`modes that are compatible with the compound (e.g.,
`chemical stability,
`thermal stability, etc.). In some
`cases, multiple modes of operation may be combined.
`The most common modes of solids generation will be
`discussed below, including thermal cooling crystalli-
`zation, anti-solvent and evaporative crystallization.
`Less common process modes include melt crystalli-
`zation, flash or quench cooling and template-directed
`crystallization. It is important to note that generation
`of maximal diversity in solid form requires multiple
`modes of operation [6,18,58].
`In thermally induced cooling crystallization, sam-
`ples created in the sample preparation process de-
`scribed above are subjected to temperature ramps.
`Prior to beginning the temperature ramp, samples are
`exposed to an elevated temperature for a short period
`of time in order to dissolve the API in the crystalliza-
`tion medium. Although dissolution can be achieved
`most simply by diffusion and convection from the
`heating process, addition of external energy can speed
`up the process (e.g., sonication). Samples may be
`optically inspected (see Fig. 1) and vessels that contain
`undissolved solids can be flagged in the database for
`further analysis. For instance, undissolved samples
`may be treated as slurry conversion experiments and
`monitored over time for crystal form changes. The
`thermal cycle is then initiated, using controlled cooling
`to induce supersaturation. In this mode of crystalliza-
`tion, samples continually experience an under cooling
`and, based on the level of supersaturation in the vessel,
`may recrystallize at a given temperature after a period
`of time. Thermal crystallization tends to generate a
`cumulative number of samples that are produced over
`time in a fashion approximating a square root function,
`as illustrated in Fig. 2. This means that initially there is
`a small bolus of ‘‘hits’’, after which the rate of
`crystallization tails off over a period of time, typically
`in days to weeks. This results in a manageable hit rate
`
`

`

`282
`
`S.L. Morissette et al. / Advanced Drug Delivery Reviews 56 (2004) 275–300
`
`methods, differential rates of solvent loss from mix-
`tures result in unknown composition of the crystalli-
`zation medium at the time of crystal nucleation. In
`addition, the degree of supersaturation changes over
`the course of the experiment, often resulting in the
`appearance of multiple crystal forms. The evaporative
`mode of solids generation typically produces through-
`put and hit rates intermediate between the thermal and
`anti-solvent processes.
`As suggested above, in appropriately configured HT
`crystallization systems, several process modes may be
`used in series or in parallel [40]. Frequently, the
`preparation of replicate plates (in some systems
`‘‘daughter’’ plates [47,51]) is necessary for parallel
`processing by different process modes. Systems may
`be additionally equipped with the ability to serially
`process sample arrays using different process modes
`[59]. This feature is particularly attractive for cases
`where only small quantities of sample are available,
`increasing the drive to generate useful information
`from every sample. Here, samples may be processed
`by optimal modes first (e.g., thermal crystallization),
`then a secondary process step can be applied to max-
`imize the hit rate. Another example where this feature is
`useful is in the case of salt selection, especially in early
`drug discovery. Upon the addition of salt forming acids
`or bases, the solubility of the compound is modulated
`by in-situ salt formation, often resulting in reduced or
`non-existent driving forces for crystallization (e.g.,
`subsaturation) of the salt species, particularly in polar
`
`Fig. 2. Typical rate of appearance of solids during a thermally driven
`HT crystallization experiment [65].
`
`Fig. 1. Photo of optical inspection station. (Inset shows close up of
`crystallization vessel that contains crystals.) (Courtesy of Trans-
`Form Pharmaceuticals, 2002.)
`
`for analysis, on the order of approximately 10% in
`aggregate. This mode of solids generation has the
`lowest throughput rate, typically, because experiments
`span days to weeks, with system residence times of
`months being possible.
`In contrast, anti-solvent addition, also known as
`‘‘crash-out’’ (or ‘‘drown out’’) crystallization, relies
`on the fact that an API is soluble to varying degrees in
`the crystallization medium, but is largely insoluble in
`a particular solvent or solvents (e.g., the anti-solvent).
`As a result, this mode of crystallization can operate at
`high-throughput rates, with samples being turned
`arou

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