throbber
1
`
`1 R
`
`elevance of Solid-state Properties
`for Pharmaceutical Products
`
`Rolf Hilfiker, Fritz Blatter, and Markus von Raumer
`
`1.1
`Introduction
`
`Many organic and inorganic compounds can exist in different solid forms [1–6].
`They can be in the amorphous (Chapter 10), i.e., disordered, or in the crystal-
`line, i.e., ordered, state. According to McCrone’s definition [2], “The polymorph-
`ism of any element or compound is its ability to crystallize as more than one
`distinct crystal species”, we will call different crystal arrangements of the same
`chemical composition polymorphs. Other authors use the term “polymorph”
`more broadly, including both the amorphous state and solvates (Chapter 15).
`Since different inter- and intramolecular interactions such as van der Waals in-
`teractions and hydrogen bonds will be present in different crystal structures, dif-
`ferent polymorphs will have different free energies and therefore different phys-
`ical properties such as solubility, chemical stability, melting point, density, etc.
`(Chapter 2). Also of practical importance are solvates (Chapter 8), sometimes
`called pseudopolymorphs, where solvent molecules are incorporated in the crys-
`tal lattice in a stoichiometric or non-stoichiometric [6, 7] way. Hydrates (Chapter
`9), where the solvent is water, are of particular interest. If non-volatile molecules
`play the same role, the solids are called co-crystals. Solvates and co-crystals can
`also exist as different polymorphs, of course.
`In addition to the crystalline, amorphous and liquid states, condensed matter
`can exist in various mesophases. These mesophases are characterized by exhibit-
`ing partial order between that of a crystalline and an amorphous state [8, 9].
`Several drug substances form liquid crystalline phases, which can be either ther-
`motropic, where liquid crystal formation is induced by temperature, or lyotropic,
`where the transition is solvent induced [10–12].
`Polymorphism is very common in connection with drug substances, which
`are mostly (about 90%) small organic molecules with molecular weights below
`600 g mol–1 [13, 14]. Literature values concerning the prevalence of true poly-
`morphs range from 32% [15] to 51% [16, 17] of small organic molecules. Ac-
`cording to the same references, 56 and 87%, respectively, have more than one
`
`Polymorphism: in the Pharmaceutical Industry. Edited by Rolf Hilfiker
`Copyright © 2006 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
`ISBN: 3-527-31146-7
`
`IPR2018-00126
`
`Page 1 of 20
`
`I-MAK 1015
`
`

`

`2
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`
`solid form if solvates are included. When a compound is acidic or basic, it is of-
`ten possible to create a salt (Chapter 12) with a suitable base or acid, and such
`a salt can in turn often be crystallized. Such crystalline salts may also exist as
`various polymorphs or solvates. Obviously, solvates, co-crystals and salts will
`have different properties from the polymorphs of the active molecule. Since
`salts generally have higher water solubility and bioavailability than the corre-
`sponding uncharged molecule, they are popular choices for drug substances.
`About half of all active molecules are marketed as salts [14, 18]. Polymorphs,
`solvates, salts, and co-crystals are schematically depicted in Fig. 1.1. We will use
`the term “drug substance” for the therapeutic moiety, which may be a solvate,
`salt or a co-crystal, while the single, uncharged molecule will be called the “ac-
`tive molecule”.
`Most drug products (formulated drug substances) are administered as oral
`dosage forms, and by far the most popular oral dosage forms are tablets and
`other solid forms such as capsules. Drugs for parenteral application are also of-
`ten stored as solids (mainly as lyophilized products) and dissolved just prior to
`use since in general the chemical stability of a molecule in the solid form is
`much higher than in solution. Drugs administered by inhalation have become
`increasingly popular, and dry powder inhalers are now commonly in use. Evi-
`dently, therefore, both the solid form of the drug substance and the selected ex-
`cipients have a strong impact on the properties of the formulated drug. Even if
`the envisaged market form of the drug is a solution, information about the sol-
`id-state properties of the drug substance may still be necessary [19]. If different
`forms have significantly different solubilities, it may be possible to unintention-
`ally create a supersaturated solution with respect to the least soluble form by
`creating a concentrated solution of a metastable form. Also, the drug substance
`will in most cases be handled as a solid in some stages of the manufacturing
`process, and its handling and stability properties may depend critically on the
`solid form.
`
`Fig. 1.1 Schematic depiction of various types of solid forms.
`
`IPR2018-00126
`
`Page 2 of 20
`
`I-MAK 1015
`
`

`

`1.1 Introduction
`
`3
`
`In fact, the whole existence of a drug is affected by the properties of the solid
`form, and the final goal of solid form development is to find and select the sol-
`id with the optimal characteristics for the intended use.
`Initially, when the drug substance is first produced, one has to be certain that
`the desired solid form is obtained in a consistent, pure and reproducible man-
`ner. Subsequently, when it is formulated to obtain the drug product, one has to
`make sure that no undesired transitions occur (Chapter 13). For this phase, a
`profound knowledge of potential solvate formation is especially useful. It is
`highly advisable to avoid using solvents that can form solvates with the drug
`substance in the formulation process. Otherwise, such solvates might be gener-
`ated during formulation and subsequently desolvated in a final drying step. In
`such a situation the final polymorph would probably differ from the initial one
`– an undesirable effect in most cases. Similarly, the energy–temperature dia-
`gram (Chapter 2) of the polymorphs and the kinetics of the change from one
`polymorph into another should be known so that one can be sure that tempera-
`ture variations during the formulation process will not lead to an unacceptable
`degree of change in the solid form.
`In the next step, when the drug substance or drug product is stored during
`its shelf-life, it is imperative that the solid form does not transform over time.
`Otherwise, important properties of the drug might change drastically. Stability
`properties have to be evaluated with respect to ambient conditions, storage, and
`packaging. Thermodynamic stability depends on the environment. A solvate, for
`example, represents a metastable form under ambient conditions but is likely to
`be the most stable form in its solvent. Thermodynamically, any metastable form
`will eventually transform into a more stable form. The kinetics under which
`this transformation occurs, however, are polymorph specific. Therefore, the exis-
`tence of a more stable polymorph does not necessarily imply that a metastable
`polymorph cannot be developed.
`In the final step, when the patient takes the drug, the solubility and dissolu-
`tion rate of the drug substance will be influenced by its solid form. This will af-
`fect the bioavailability if solubility is a rate-limiting step, i.e., if the drug belongs
`to class 2 or 4 of the biopharmaceutics classification system (BCS) [20]. Because
`a change of solid form may render a drug ineffective or toxic, regulatory autho-
`rities demand elucidation and control of solid-state behavior (Chapter 15).
`Finally, thorough, experimentally obtained knowledge of the solid-state behav-
`ior also has the advantages that a good patent situation for a drug substance
`can be obtained and that valuable intellectual property can be generated (Chap-
`ter 14). Although in hindsight everything may appear to be easy and straightfor-
`ward, crystalline molecular solid-state forms are non-obvious, novel and require
`inventiveness. For instance, typically, many attempts to crystallize an amor-
`phous drug substance fail until, suddenly, a stable crystalline form is obtained.
`Once seed crystals are available, the crystallization becomes the simple last step
`of a production process.
`
`IPR2018-00126
`
`Page 3 of 20
`
`I-MAK 1015
`
`

`

`4
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`
`1.2
`Drug Discovery and Development
`
`Typically, it takes eight to twelve years, or sometimes even longer, for a mole-
`cule with biological activity to progress from its first synthesis to market intro-
`duction as an efficacious, formulated drug [21]. This process is normally divided
`into two main phases: (a) research or discovery and (b) development [22]. In the
`research phase, the appropriate target for a particular disease model is identified
`and validated, and candidate molecules are synthesized or chosen from libraries.
`They are primarily tested with respect to binding affinity to the target or, if pos-
`sible, directly for their potential to alter a target’s activity. Sometimes other pa-
`rameters, such as selectivity, are also considered. Promising candidates are
`usually termed “hits”. As a rule at this stage, limited attention is paid to the
`possibility to formulate a drug for a certain administration route. Often, from a
`drug delivery aspect, simple vehicles like DMSO solutions are used. As a result,
`the activity of especially poorly water-soluble drugs may not be identified at all
`because they precipitate under the used in vitro conditions [23]. In a medicinal
`chemistry program the “hits” are then modified to improve physicochemical pa-
`rameters such as solubility and partition coefficient. This is the first time that
`solid-state properties come into play. When solubility is evaluated, it is critical to
`know whether the solubility of an amorphous or crystalline substance was mea-
`sured. Permeation measurements are performed using, e.g., Caco-2 [24], PAM-
`PA [25] or MDCK [26] assays, and dose–response studies are conducted in in vi-
`tro models. Selectivity is assessed in counter screens. At the same time, prelim-
`inary safety studies are carried out, and IP opportunities are assessed. Struc-
`ture–activity relationship (SAR) considerations play a large role at this stage.
`Molecules that show promise in all important aspects are called “leads”. Often
`several series of leads are identified and are then further optimized and scruti-
`nized in more sophisticated models, including early metabolic and in vivo stud-
`ies. Both pharmacokinetics (PK, the quantitative relationship between the admi-
`nistered dose and the observed concentration of the drug and its metabolites in
`the body, i.e., plasma and/or tissue) and pharmacodynamics (PD, the quantita-
`tive relationship between the drug concentration in plasma and/or tissue and
`the magnitude of the observed pharmacological effect) are studied in animal
`models to predict bioavailability and dose in humans. Simultaneously with char-
`acterization of the drug substance, a proper dosage form needs to be designed,
`enabling the drug substance to exert its maximum effect. For freely water-solu-
`ble drugs this is less critical than for poorly water-soluble drugs, which without
`the aid of an adequate dosage form cannot be properly investigated in the re-
`search stage. In the discovery phase, high-throughput methods play an increas-
`ingly important role in many aspects, such as target identification, synthesis of
`potential candidate molecules, and screening of candidate molecules. Consider-
`ing that only about 1 out of 10 000 synthesized molecules will reach the market
`[21], high-throughput approaches are a necessity. The optimal molecule arising
`from these assessments is then promoted to the next stage, i.e., development.
`
`IPR2018-00126
`
`Page 4 of 20
`
`I-MAK 1015
`
`

`

`1.2 Drug Discovery and Development
`
`5
`
`Fig. 1.2 Drug development process with a description of respective phases,
`approximate number of test persons, timelines and attrition rates. These
`numbers are a rough guideline only and can differ significantly according
`to the specific indication, the characteristics of the drug substance, etc.
`
`The development process of a pharmaceutical product is depicted in Fig. 1.2. It
`consists of a non-clinical and a clinical phase. While drug companies’ approaches
`to the non-clinical phase can differ somewhat, the clinical phase is treated very
`similarly due to regulatory requirements. In the non-clinical phase enough data
`is gathered to compile an Investigational New Drug Application (IND) in the
`US or a Clinical Trial Application (CTA) in the European Union, which is the pre-
`requisite for the first use of the substance in humans. For obvious reasons, partic-
`ular emphasis is placed on toxicology studies during this phase, including assess-
`ment of toxicity by single-dose and repeated-dose administration and evaluation of
`carcinogenicity, mutagenicity and reproductive toxicity. An absolute necessity at
`this stage is that the drug is maximally bioavailable, resulting in sufficient expo-
`sure of the animals to the drug to obtain an adequate assessment of its toxicity
`profile. Whenever possible, the need for animal studies is reduced by using,
`e.g., human cell in vitro tests. The non-clinical development phase lasts between
`one and two years, and the attrition rate is ca. 50% (Fig. 1.2). At the end of the
`non-clinical phase, the decision has to be made whether the neutral molecule, a
`salt, or a co-crystal will be developed. If a salt form or co-crystal is chosen, it
`has to be clear which salt (Section 1.4.1) or co-crystal is optimal. In the clinical
`phases the product is first tested on healthy volunteers and then on small and
`large patient populations. For certain disease indications, like oncology, Phase I
`studies are performed directly on patients. Approximate population sizes are given
`in Fig. 1.2. One has to bear in mind, however, that these numbers depend signif-
`icantly on the indication the drug is intended to treat. Attrition rates during the
`clinical phases are between 80 and 90%. During the clinical phases, analytical,
`process and dosage-form development continues in parallel with long-term toxi-
`cology studies. Of course, solid-state properties continue to play a crucial role dur-
`
`IPR2018-00126
`
`Page 5 of 20
`
`I-MAK 1015
`
`

`

`6
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`
`ing both chemical development of the drug substance and pharmaceutical devel-
`opment of the dosage form.
`
`1.3
`Bioavailability of Solids
`
`An issue that has to be addressed for every drug product, and which is closely
`related to its solid-state properties, is whether its solubility and dissolution rate
`are sufficiently high. This leads to the question of what the minimal acceptable
`solubility and dissolution rates are.
`Bioavailability essentially depends on three factors: solubility, permeability
`and dose [27], and the question of minimal acceptable solubility can only be an-
`swered if the other two factors are known. According to the BCS a drug sub-
`stance is considered highly soluble when the highest strength dosage is soluble
`in 250 mL of aqueous media over the pH range 1.0–7.5 [28].
`A valuable concept for estimating what the minimum solubility of a drug sub-
`stance for development purposes should be uses the maximum absorbable dose
`(MAD) [29, 30]. MAD corresponds to the maximum dose that could be absorbed
`if there were a saturated solution of the drug in the small intestine during the
`small intestinal transit time (SITT & 270 min). The bioavailable dose is smaller
`than MAD due to metabolism of components in the portal blood in the liver
`(first pass effect) and in the intestinal mucosal tissue [20]. MAD can be calcu-
`lated from the solubility, S, at pH 6.5 (corresponding to typical conditions in
`the small intestine), the transintestinal absorption rate (Ka), the small intestinal
`water volume (SIWV & 250 mL) and the SITT.
`
`MAD (mg) = S (mg mL–1) ´ Ka (min–1) ´ SIWV (mL) ´ SITT (min)
`
`(1)
`
`Human Ka can be estimated from measured rat intestinal perfusion experi-
`ments [30, 31]. It is related to the permeability (P) through SIWV and the effec-
`tive surface of absorption (Sabs) [20].
`
`Ka (min–1) = P (cm min–1) ´ Sabs (cm2)/SIWV (mL)
`
`(2)
`
`In the absence of active diffusion, permeability is related to the diffusion coeffi-
`cient (D), the partition coefficient K (= cin membrane/cin solution) and the mem-
`brane thickness (d).
`
`P (cm min–1) = D (cm2 min–1) ´ K/d (cm)
`
`(3)
`
`In reality, proportionality between the partition coefficient and the permeability
`is only found for a rather small range of partition coefficients [24, 32]. This is
`because the model of a single homogeneous membrane is an oversimplification.
`The intestinal wall is better represented by a bilayer membrane consisting of an
`
`IPR2018-00126
`
`Page 6 of 20
`
`I-MAK 1015
`
`

`

`1.4 Phases of Development and Solid-state Research
`
`7
`
`aqueous and an adjoining lipid region. Therefore, for highly lipophilic sub-
`stances, the water layer becomes the limiting factor and leads to a decrease in
`permeability as K is increased [33].
`Implicit in Eq. (1) is that the solution stays saturated during the SITT and
`therefore that there is a large excess of solid drug in the small intestine. In de-
`riving this equation as a limiting case, the authors [29] took into account the
`dissolution kinetics of a polydisperse powder and showed how the percentage of
`the dose that is absorbed is influenced by solubility, particle size and permeabil-
`ity. They showed that for highly soluble drugs, as defined above, the percentage
`of dose absorbed is only limited by permeability. For smaller solubilities, the
`dissolution rate and hence the particle size become important factors as well.
`The influence of particle size is greatest for low-solubility and low-dose drugs.
`MAD readily translates into minimal acceptable solubility [30].
`
`Minimal acceptable solubility = S ´ {target dose (mg)/MAD}
`= target dose/{Ka ´ SIWV´ SITT}
`
`(4)
`
`Realistic values for Ka lie between 0.001 and 0.05 min–1 and vary over a much nar-
`rower range than typical solubilities (0.1 lg mL–1 to 100 mg mL–1) [30]. Consider-
`ing these facts and assuming a typical dose of 70 mg, i.e., 1 mg kg–1, minimal ac-
`ceptable solubilities between 20 lg mL–1 and 1 mg mL–1 are obtained. When mak-
`ing these estimates, one has to keep in mind that the assumptions of the model
`break down if there is possible absorption in other parts of the gastrointestinal
`tract or if the diffusivity of the drug is changed due to the meal effect, etc. [34].
`Furthermore, it is important to realize that S represents a “kinetic” solubility. A
`weakly basic drug might be freely soluble in the stomach while its equilibrium sol-
`ubility in the small intestine might be very low. Nevertheless, it may remain in the
`supersaturated state in the small intestine, in which case that “kinetic” solubility
`would be the relevant one for calculating the MAD.
`
`1.4
`Phases of Development and Solid-state Research
`
`Normally, solid-state research and development involves the following stages,
`which may also overlap:
`· deciding whether the uncharged molecule or a salt should be developed;
`· identifying the optimal salt;
`· identifying and characterizing all relevant solid forms of the chosen drug
`substance;
`· patenting new forms;
`· choosing a form for chemical and pharmaceutical development;
`· developing a scalable crystallization process to obtain the desired form of the
`drug substance;
`
`IPR2018-00126
`
`Page 7 of 20
`
`I-MAK 1015
`
`

`

`8
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`· developing a method to determine the polymorphic purity of the drug sub-
`stance;
`· formulating the drug substance to obtain the drug product;
`· developing a method to determine the polymorphic purity of the drug sub-
`stance in the drug product.
`
`Not all of these stages may be necessary for every drug substance, and the order of
`the stages may be varied according to the specific properties and behavior of the
`drug. Particularly for drugs that are poorly water soluble, polymorphism in formu-
`lations can play a crucial role since it could significantly influence the dissolution
`rate and degree of dissolution required to achieve adequate bioavailability.
`
`1.4.1
`Salt Selection
`
`Clearly, the first decision is whether it is more desirable to develop the un-
`charged molecule or, if possible, a salt thereof (Chapter 12). In general, salt for-
`mation will be possible if the molecule contains acidic or basic groups, which is
`the case for most active molecules. Since making a salt will normally involve an
`additional step in the synthesis and since the molecular weight of a salt will al-
`ways be higher than that of the neutral molecule, salts will only be chosen if
`they promise to have clear advantages compared with the free acid/base. As a
`rule, a salt is chosen if the free acid/base has at least one of the following unde-
`sirable properties:
`· very low solubility in water;
`· apparently not crystallizable;
`· low melting point (typical cutoff 80 8C [35]);
`· high hygroscopicity;
`· low chemical stability, etc.;
`· IP issues.
`
`Low water solubility is relative and always has to be assessed in the context of
`dose and permeability (Section 1.3). A very low water solubility may mean a
`high lipophilicity, enabling efficient passage through membranes, or a very
`large binding constant with the receptor, allowing a low dose. Also, the amor-
`phous state of a neutral molecule may be the best option to get high oral bio-
`availability, provided the amorphous form can be kinetically stabilized over a
`reasonable time scale. Therefore, the decision to develop a salt should be based
`on a head-to-head broad comparison, taking into consideration both in vivo per-
`formance and physicochemical properties. If the decision has been made to de-
`velop a salt, it is obviously important to carry out a broad salt screening and salt
`selection process to identify the optimal salt. Potential counterions are chosen
`based on pKa differences, counterion toxicity (preferably GRAS status [18, 36]),
`etc. (Chapter 12). Desirable properties of the salts include crystallinity, high
`water solubility, low hygroscopicity, good chemical stability, and high melting
`
`IPR2018-00126
`
`Page 8 of 20
`
`I-MAK 1015
`
`

`

`1.4 Phases of Development and Solid-state Research
`
`9
`
`Fig. 1.3 Rough guideline of when the various issues related to solid-state
`properties generally should be taken care of in the development process.
`Depending on company policies, obtained results and other circumstances,
`large shifts are possible. In particular, certain steps may have to be
`repeated due to unanticipated experimental results.
`
`point. The relative importance of these properties may vary from project to pro-
`ject. At this stage it also has to be decided whether co-crystals are to be consid-
`ered. Co-crystals can offer valuable alternatives, especially for very weak bases or
`acids. Very often, salt screening and salt selection are performed in stages: first
`a large number of salts is produced on a microscopic scale and characterized
`with a limited number of methods (e.g., birefringence, Raman, XRD) to identify
`a few promising candidates, which are then produced on a scale of a few
`100 mg and characterized in more detail.
`Different companies perform salt screening in different phases of development.
`Some even move the salt selection process to the research phase [35], but clearly
`the decision on the salt form should ideally be made no later than the beginning of
`the long-term toxicology studies, i.e., at the start of Phase I (Fig. 1.3).
`
`1.4.2
`Polymorph Screening
`
`The objective of the next important step with respect to solid-state development is
`identifying all relevant polymorphs and solvates (Chapter 11), characterizing them
`(Chapters 3 to 7), and choosing the optimal form for further chemical and phar-
`maceutical development. In the absence of solvents and humidity, the thermody-
`namically stable polymorph is the only one that is guaranteed not to convert into
`another polymorphic form. This is why this form is most often chosen for the
`drug product [31]. The disadvantage of the thermodynamically stable form is, of
`course, that it is always the least soluble polymorph (Chapter 2) and therefore
`has the lowest bioavailability. But in most cases this is a small price to pay for
`the very large advantage of absolute kinetic stability. Differences in the solubility
`of various polymorphs are typically lower than a factor of 2 (see Ref. [37] for a re-
`
`IPR2018-00126
`
`Page 9 of 20
`
`I-MAK 1015
`
`

`

`10
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`
`view of literature data), but sometimes as much as a five-fold difference can be ob-
`served [38]. In cases where several enantiotropically related forms exist and where
`the transition temperature is around room temperature, the choice may be diffi-
`cult, but it is based on the same criteria as for all solid forms. The kinetics of in-
`terconversion from one form into the other and the reproducibility of producing
`consistently the same ratio of polymorphs are important.
`Apart from the thermodynamically stable polymorph of a drug substance, hy-
`drates are also very popular components of the final dosage form. Owing to the
`ubiquity of water vapor, hydrates are often the thermodynamically stable form
`at ambient conditions. If a certain hydrate is stable within a rather large range
`of humidities, it may therefore be much easier to formulate the hydrate in a
`controlled way and to subsequently store and package it.
`In a few cases, a metastable form might be preferable [31], normally for one
`of the following reasons:
`
`too low a solubility (and bioavailability) of the stable form;
`1.
`2. high dissolution rate needed for quick-relief formulations;
`3. manufacturing difficulties;
`4.
`IP issues;
`5.
`chemical instability of the thermodynamically stable form due to
`topochemical factors.
`
`(1) If the solubility of the stable polymorph is critically low (Section 1.3) and no
`salt is feasible, several options exist [39]. Liquid-like formulations (emulsions,
`microemulsions, liposomal formulations) or soft gelatin capsules filled with
`solutions of the drug in a non-aqueous solvent may be used. Alternatively,
`a metastable solid form, a solvate or a co-crystal might be selected for devel-
`opment. If a solid form with a higher solubility than the thermodynamically
`stable form is desired, it is often better to use the amorphous form rather than
`a metastable polymorph, provided that the glass transition temperature (Tg) of
`the amorphous form is sufficiently high (Chapter 10) [40]. Firstly, the amor-
`phous form often has a ten-fold or higher increased solubility relative to
`the stable form [41], while metastable polymorphs typically have a less than
`a two-fold higher solubility, as mentioned above. Secondly, it is normally im-
`possible to stabilize a metastable form reliably by excipients, since they can
`only interact with the surface of the crystals of the metastable drug substance.
`This will change the surface free energy, but for crystal sizes larger than some
`tens of nanometers, the contribution of the surface free energy to the total
`free energy is negligible. The best way to stabilize a metastable form kineti-
`cally is to ensure the absence of any seeds of the stable form because such
`seeds have a very large effect on the kinetics of transformation [42]. The amor-
`phous form, however, can be stabilized, for example, by creating a solid dis-
`persion with a polymer [43, 44]. Such a dispersion will be highly kinetically
`stable if two conditions are fulfilled: if it remains in the glassy state under
`the storage conditions, thus blocking all translational diffusion, and if the
`
`IPR2018-00126
`
`Page 10 of 20
`
`I-MAK 1015
`
`

`

`1.4 Phases of Development and Solid-state Research
`
`11
`
`drug substance molecules are molecularly dispersed within the matrix. In any
`case, irrespective of whether a crystalline or disordered metastable form is to
`be developed, very careful kinetic stability studies will be necessary. For amor-
`phous solids, particular attention has to be paid to the lowering of the glass
`transition temperature due to humidity.
`
`(2) In some instances, quick onset of action of a drug is of particular impor-
`tance. In such cases, metastable forms with a higher dissolution rate may
`accelerate the uptake of the drug and may therefore act faster.
`
`(3) Different polymorphs will also have different mechanical properties, such
`as hardness, powder flow properties, compressibility and bonding strength.
`A well-known example is acetaminophen (also known as paracetamol),
`where the thermodynamically stable form (monoclinic form I) cannot be
`compressed into stable tablets while the metastable form II (orthorhombic)
`can as it shows more favorable properties with respect to plastic deforma-
`tion [45]. In very rare cases, this might lead to a decision to develop a meta-
`stable form.
`
`(4) If the thermodynamically stable polymorph is protected by patents, while
`other forms are free, the respective drug substance can be marketed as a
`metastable form without obtaining a license from the patent owner (Chap-
`ter 14) [5].
`
`(5) Generally, the thermodynamically most stable polymorph is also the most
`stable chemically (Chapter 2) [31]. This has been attributed to the fact that
`its density is typically higher, but it could also be explained by its lower free
`energy. Only in extremely rare cases, where the arrangement of atoms in
`the stable polymorph favors an intermolecular chemical reaction, could its
`chemical stability be lower. In such cases, development of a metastable
`form might be advisable.
`
`A very important question is, of course, when a polymorphism screening should
`be carried out and when the choice of form to develop should be made. Since dif-
`ferent solid forms have different properties and may have different bioavailabil-
`ities, it is definitely advisable to select the final form together with the accompany-
`ing formulation before carrying out pivotal clinical studies [19, 46]. It is, therefore,
`critical to have at least identified the thermodynamically stable form along with
`important hydrates by the end of Phase I at the latest (Fig. 1.3). Accordingly, by
`that time a polymorphism screening that is primarily designed to identify these
`forms with a large probability should have been completed. Owing to economic
`reasons and the expected attrition rate of up to 90% of potential drug candidates
`after this stage, a full polymorphic screening, which identifies all relevant meta-
`stable forms as well, may need to be deferred. However, this should only be the
`exception because knowledge of metastable phases, thermodynamic stability as
`a function of temperature and conditions for solvate formation is crucial for the
`design of crystallization and formulation processes.
`
`IPR2018-00126
`
`Page 11 of 20
`
`I-MAK 1015
`
`

`

`12
`
`1 Relevance of Solid-state Properties for Pharmaceutical Products
`
`While the kinetic stability of dry metastable forms is not much influenced by
`additives, as mentioned above, additives and impurities can influence their ki-
`netic stability in solutions and suspensions [47] by affecting both nucleation
`and growth rates. Therefore, a polymorphism screening that is performed with
`an early batch of drug substance still containing many impurities may provide
`different results from a screening performed with a later, purer batch. In partic-
`ularly unfortunate cases, important forms may not be discovered in the initial
`screening. Therefore, it is highly advisable to repeat at least a limited poly-
`morphism screening with a batch of drug substance produced with the final
`GMP procedure, which has the impurity profile of the product to be marketed.
`Clearly, the unexpected appearance of a new form at a late stage can be disas-
`trous. A very well publicized example is that of ritonavir (Norvir) [38, 48]. When
`it was launched on the market, only form I was known. One marketed formula-
`tion consisted of soft gelatin capsules filled with a nearly saturated solution of
`form I. About two years after market introduction, some capsules failed the dis-
`solution test due to precipitation of a new, thermodynamically more stable form
`of ritonavir (form II). The solubility difference between forms I and II is about
`a factor of 5 [38], which is unusually high. In the end, the original formulation
`had to be taken off the market, and a new formulation had to be developed with
`considerable effort and expense [38]. While this is certainly an extreme case,
`there are many instances of new polymorphs appearing in Phase II and Phase
`III studies, leading to considerable difficulties [49].
`
`1.4.3
`Crystallization Process Development
`
`After selecting the appropriate solid form for

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