throbber
6.2
`
`ROLE OF PREFORMULATION IN
`
`DEVELOPMENT OF SOLID
`
`DOSAGE FORMS
`
`OMATHANU P. PERUMAL AND SATHEESH K. PODARALLA
`
`South Dakota State University, Brookings, South Dakota
`
`Contents
`
`6.2.1
`
`Introduction
`
`6.2.2
`
`6.2.3
`
`6.2.4
`
`6.2.5
`
`Physical/Bulk Characteristics
`6.2.2.1 Crystallinity and Polymorphism
`6.2.2.2 Hydrates/Solvates
`6.2.2.3 Amorphates
`6.2.2.4 Hygroscopicity
`6.2.2.5 Particle Characteristics
`
`6.2.2.6 Powder Flow and Compressibility
`
`Solubility Characteristics
`6.2.3.1
`pKa and Salt Selection
`6.2.3.2 Partition Coefficient
`
`6.2.3.3 Drug Dissolution
`6.2.3.4 Absorption Potential
`
`Stability Characteristics
`6.2.4.1
`Solid—State Stability
`6.2.4.2 Solution-State Stability
`6.2.4.3 Drug—Excipient Compatibility
`Conclusions
`
`References
`
`Pharmaceutical Manufacturing Handbook: Production and Processes, edited by Shayne Cox Gad
`Copyright © 2008 John Wiley & Sons, Inc.
`
`933
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 1 of 43)
`
`

`
`934
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`6.2.1
`
`INTRODUCTION
`
`The advent of combinatorial chemistry and high—throughput screening (HTS) has
`exponentially increased the number of compounds synthesized and screened during
`the drug discovery phase. However, the overall efficiency of the drug discovery
`process is still exceedingly low (only 1 in 10,000 makes it to the market). Drug dis-
`covery is mostly driven by “activity screens” with little emphasis on “property
`screens.” This is exemplified by the fact that 40% of attrition in drug discovery and
`development is attributed to poor biopharmaceutics and pharmacokinetic proper-
`ties [l], which in turn are related to poor physicochemical properties. As a result,
`pharmaceutical companies have started to redesign their strategies by including
`property screens quite early in the discovery stage
`Preformulation is the study
`of fundamental properties and derived properties of drug substances. In other
`words, preformulation is the first opportunity to learn about the drug’s physico-
`chemical properties from the perspective of transforming a biologically active mol-
`ecule to a “druggable” molecule. The type and extent of preformulation activities
`vary in a discovery and generic setting.
`The main goal of a drug discovery program is to develop an orally deliverable
`molecule for obvious reasons of ease of manufacture and convenience of drug
`administration. More than 75% of the drug products in the market are oral formula-
`tions, of which more than half are solid dosage forms
`The “rule of five” devel-
`oped by Christopher Lipinski [4] is one of the “physicochemical screens” to weed
`out molecules with poor physicochemical properties very early in the drug discovery
`process. According to Lipinski’s rule, a drug will show poor oral absorption if it does
`not conform to any of the two physicochemical requirements listed in Table 1. The
`rule of five is applicable only to small molecules and it relates the chemical proper-
`ties of the drug to its solubility and permeability characteristics. During the initial
`stages of drug discovery, the preformulation activities are mainly focused on devel-
`oping a water—soluble compound for early activity studies and preclinical testing in
`animals. At this stage, the preformulation scientist is faced with the challenge of
`working with a limited quantity of compound (few milligrams) for testing a long list
`of physicochemical parameters. On the other hand, developing preclinical formula-
`tions can be quite a daunting task given the fact that toxicological studies require
`a high dose of drug (l0—l00 times above the effective dose) to be delivered in a
`small volume of the formulation. Preformulation activities increase as the molecule
`
`proceeds through the development phase. The “discovery and development phar-
`
`TABLE 1 Lipinski Rule of Five for Orally Active Compounds
`
`Physicochemical Parameter
`
`Lipinski rule
`
`Molecular weight
`log P
`Hydrogen bond donors
`
`Not more than 500Da
`Not more than 5
`
`Not more than 5 hydrogen bond donors expressed as the
`sum of OH’s and NH’s
`
`Hydrogen bond acceptors
`
`Not more than 10 expressed as the sum of OH’s and NH’s
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 2 of 43)
`
`

`
`PHYSICAL/BULK CHARACTERISTICS
`
`935
`
`maceutics” documentation forms a significant portion of the investigational new
`drug application (IND) application and new drug application (NDA) filed to the
`U.S. Food and Drug Administration (FDA). In a generic setting, preformulation
`studies are mainly focused on developing a formulation that is bioequivalent to the
`innovator’s product with the main objective of filing an abbreviated new drug appli-
`cation (ANDA). A strong preformulation team can generate intellectual property
`in the form of new salts, solid—state forms, or new stabilized formulations of the drug
`for an innovator and/or a generic manufacturer.
`In the present chapter, the discussion is mainly focused on preformulation testing
`for oral solid dosage forms in a drug discovery setting. The chapter address the fol-
`lowing goals of preformulation:
`to gain knowledge about the physicochemical
`characteristics of the drug, (ii) to define the physical characteristics of the drug, (iii)
`to understand the stability characteristics of the drug, and (iv) to determine the
`compatibility of eXcipients with the drug. In this chapter, we have grouped all
`the parameters under three sections and discussed in a logical sequence for the
`convenience of the reader.
`
`6.2.2 PHYSICAL/BULK CHARACTERISTICS
`
`The bulk or physical characteristics of a drug substance are mainly dictated by its
`solid—state properties. Purity of the drug substance is a fundamental property that
`is characterized at the beginning of preformulation studies. In the initial stages of
`drug development, the drug is usually not very pure. Nevertheless, it is essential to
`know the purity of the material at hand using simple measurements such as melting
`point. This would serve to set drug specifications during later stages of drug develop-
`ment. Differential scanning calorimetry (DSC) requires very little sample (1-5 mg)
`and is a useful tool to estimate the purity of the compound. The drug sample is
`heated in a crucible, where the difference in heat between the sample and a refer-
`ence crucible is seen as an endotherm or eXotherm in the thermogram depending
`on whether heat is taken up or given up, respectively, by the sample. The integrated
`area under the endotherm or eXotherm gives a measure of the heat or enthalpy
`involved in this process. Melting is seen as an endothermic event and the purity of
`the sample will govern the peak position, shape, sharpness, and heat of fusion (AHf).
`DSC is sensitive in detecting impurities to the extent of 0.002 mol %
`The DSC
`findings should be substantiated by a stability—indicating high—performance liquid
`chromatography (HPLC) assay. On the other hand, thin—layer chromatography
`(TLC) may be used to qualitatively detect the number of impurities in the drug
`sample. Impurity profiling is an important aspect of the drug development process,
`particularly for optimizing the synthetic process. The impurities can originate from
`many sources, including starting materials, intermediates, synthetic processes, or
`degradation reactions
`The regulatory guidelines stipulate that any impurity
`>0.05% of total daily dose (for drugs with a dose <2 g/day) or >0.l5% of total daily
`dose (for drugs with a dose >2 g/day) should be evaluated for its safety
`Organo—
`leptic properties such as color, taste, and odor are assessed qualitatively to set bulk
`drug specifications. If the drug has an unacceptable taste or odor, the chemistry
`group is advised to make a suitable salt form of the drug.
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 3 of 43)
`
`

`
`936
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`6.2.2.1 Crystallinity and Polymorphism
`
`The majority of the drugs exist in crystalline form and are characterized by their
`crystal habit and crystal lattice. The crystal habit describes the external morphology
`of the crystal, including shape and size, while the crystal lattice describes the internal
`arrangement of molecules in the crystal (Figures 1a and b). Drug molecules arrange
`in more than one way in a crystal, and this difference in the internal arrangement
`of crystals is known as polymorphism. The polymorphs have the same elemental
`composition but differ in their physical, chemical, thermodynamic, stability, and
`spectroscopic properties. A crystal lattice represents the space in which molecules
`arrange in different ways. Organic molecules arrange in one or more of the seven
`possible crystal systems: triclinic, monoclinic, orthorhombic, tetragonal, rhombohe—
`dral, hexagonal, and cubic
`Each crystal system is characterized by its three-
`dimensional geometry and angles between the different crystal faces. The crystal
`lattice geometry is obtained using single—crystal X—ray diffractometry (XRD) and
`the details can be found elsewhere
`The difference in the crystal lattice of a drug
`arises as a result of the difference in packing of the molecules if the molecules are
`conformationally rigid (e.g., chlordiazepoxide) or due to the differences in confor-
`mation for flexible drug molecules (e.g., piroxicam). Although polymorphs differ in
`their internal crystal lattice, it may not be necessarily reflected in their external
`crystal habit (Figure 1b). In other words, a drug can exist in different crystal habits
`without any change in the internal crystal lattice (isomorphs).
`For example,
`Crystal habit is mainly dependent on crystal growth conditions
`Figure 1a shows two different crystal habits for a given crystal lattice. A prismatic
`crystal habit will result if the growth is equal in all directions, while plates are formed
`if the growth is slow in one direction. Alternatively, needle—shaped crystals (acicular)
`are formed when the growth is slow in two directions. Thus, the crystal habits can
`
`(6)
`
`(b)
`
`(0)
`
`FIGURE 1 Schematic of crystal habits, polymorphs and amorphous drug forms. (a) Two
`crystal habits are shown. The internal crystal lattice is the same while the external morphol-
`ogy is different. (b) In a crystal the molecules are arranged in a regular fashion. However,
`the arrangement may vary depending on how the molecules orient themselves in the internal
`crystal lattice. The internal crystal lattice is different in all the three polymorphic forms. The
`polymorphs may or may not differ in their external morphology. (c) Random arrangement
`of molecules in amorphous form.
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 4 of 43)
`
`

`
`PHYSICAL/BULK CHARACTERISTICS
`
`937
`
`be altered without any change in the internal crystal lattice by varying the crystalli-
`zation conditions. The polarity of the crystallizing solvent mainly influences the
`crystal habit by preferentially adsorbing to one surface of the crystal face. Similarly,
`surfactants or additives are added to the crystallization medium to prevent or
`promote the growth of a specific crystal habit
`Crystal habits mainly differ in
`physicomechanical properties such as packing, flow property, compressibility, and
`tablettability. Acetaminophen crystallizes as polyhedral crystals when crystallized
`from water and as plates when crystallized from ethanol—water (Figure 2a). Both
`these crystal habits are isomorphic [9], that is, have the same internal crystal arrange-
`ment, since their melting points and heats of fusion were similar (melting point
`178 “C and Mt‘
`l77kcal/mol). The polyhedral crystals have better flow and
`
`In[1/(1—D)]
`
`Thin platelike erystals
`
`— Polyhedral crystals
`
`O 10 20 3040 50 60 70 80
`
`Compression pressure (MPa)
`(b)
`
`FIGURE 2 Difference in crystal habit of acetaminophen and resultant difference in com-
`pressibility (a) Acetaminophen crystallizes as either platy crystals or polyhedral crystals
`depending on the solvent of crystallization. Both crystal habits have the same internal crystal
`lattice since they showed the same melting point. (b) Difference in compression behavior of
`two crystal habits. The x axis represents the compression pressure while the y axis represents
`the densification of the drug sample on compression. This plot is known as Heckel plot. The
`polyhedral crystal habit shows a higher densification implying better compressibility than
`plate like crystals. [From Garekani, H. A., Ford, J. L., Rubinstein, M. H., and Raj abi—Sahboomi,
`A. R., International Journal of Pharmaceutics, 187, 77-89, 1999. Reproduced with permission
`from Elsevier.)
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 5 of 43)
`
`

`
`938
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`compression properties than platy crystals, which were brittle and fragmented
`during tableting (Figure 2b). Crystal habits are characterized using optical and
`electron microscopy, but their internal crystal lattice should be confirmed using
`DSC, XRD, and spectroscopic techniques.
`Polymorphs are generated by crystallizing the drug from various solvents. The
`solvents are usually those that are used in the synthesis and purification of the bulk
`drug but may also include solvents used in drug formulations [10]. By convention,
`polymorphs are named based on their order of discovery, such as forms I, II or A,
`B or 0L, B. In general form I is considered the most stable and least soluble form,
`while form II is considered the more soluble and least stable form. The least stable
`
`and more soluble polymorphic form is usually called the metastable form. They are
`not “unstable” but “metastable,” because the least stable form can remain stable
`
`provided the conditions are controlled to prevent its conversion to the more stable
`polymorphic form. Polymorphs are characterized by their solubility and stability
`differences with respect to temperature (Figure 3). Thermodynamically, polymorphs
`are classified as enantiotropic or monotropic depending on their thermal revers-
`ibility from one form to another [11]. Enantiotropic polymorphs are reversible
`polymorphs, where one form (form I) is more stable at higher temperature, while
`the other form (form II) is stable at lower temperature. They are characterized by
`a transition point (TS) below the melting points of both forms (Figure 3a). The
`transition point represents the temperature at which one form converts to another.
`In the temperature—solubility curve, this is represented by the intersection of the
`solubility curves of both forms; that is, at the transition temperature, both poly-
`morphs have the same solubility. As shown in Figure 3a, form II can convert to form
`I at a temperature above TS, while form I can convert to form II at a temperature
`below TS. On the other hand, monotropic polymorphs are not reversible but can
`only convert from the metastable form to the stable form. Here, TS is higher than
`the melting point of both forms (Figure 3c). Both forms are stable in the entire
`temperature range below T5.
`The different polymorphs are generated based on their solubility differences in
`a given solvent. According to Ostwald’s rule [12], the least stable or highly energetic
`form (form II, or metastable) will precipitate out first from a supersaturated solu-
`tion followed by the stable or less energetic form (form I). Supersaturation is
`achieved by antisolvent addition or by altering the temperature. So, if the initial
`precipitate is separated rapidly, it would have predominantly the metastable form.
`Alternatively, the stable form can be melted and rapidly cooled to crystallize the
`metastable form. A stable or metastable polymorphic form is also used as a “seed”
`to preferentially grow and isolate the desired form during drug crystallization [13].
`Several rules have been proposed to differentiate enantiotropic and monotropic
`polymorphs [11, 13]. A simple way to differentiate enantiotropic and monotropic
`polymorphs is the use of the heat—cool cycle in DSC [11]. As shown in Figure 3b,
`the enantiotropic polymorph is characterized by the appearance of solid—solid
`endothermic transition of form II to I followed by melting of form I. On cooling
`the melt of form I followed by reheating, the same thermogram is regenerated,
`proving the reversibility of the polymorphs. In monotropic polymorphs (Figure 3d),
`the thermogram is characterized by melting of metastable (form II) and recrystal-
`lization to form I followed by melting of form I. On cooling and reheating the
`sample, the transition and recrystallization peaks are not seen, indicating the irre-
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 6 of 43)
`
`

`
`PHYSICAL/BULK CHARACTERISTICS
`
`939
`
`Temperature (°C)
`(C)
`
`I Endotherm
`
`—> Second heat
`cycle
`
`4: Cool Cycle
`
`Tcrys
`
`Tm,||
`
`:> First heat c cle
`Tm,|
`y
`
`
`Temperature (°C)
`(d)
`
`E
`In ‘
`2
`O
`<1)
`
`
`
`5
`=
`‘g
`I
`
`Temperature (°C)
`(a)
`
`—>Second heat
`Cycle
`
`4: Cool cycle
`
`:> First heat c cle
`Tm,|
`y
`
`
`l Endotherm
`
`
`
`Ts
`
`Temperature (°C)
`(b)
`
`E
`In
`2
`O
`<1)
`
`2
`2
`E
`E
`
`FIGURE 3 Difference between enantiotropic and monotropic polymorphs. (a) Solubility
`of enantiotropic polymorphs as function of temperature. The dotted line indicates the melting
`curve. Form 1 is less soluble below the transition temperature (Ts), while form 11 is more
`soluble above Ts. Form 1 has a higher melting point (Tm,I) than form 11 (Tm,II). Below Ts,
`form I is converted to form 11 and above Ts form 11 coverts to form I. (b) Thermogram gen-
`erated from heat—cool—heat cycle in DSC. In the first heating cycle two endotherms are seen
`corresponding to conversion of form 11 to form I and melting of form 1, respectively. On
`cooling both events show up as exotherms and on second heating cycle both endotherms
`reappear, indicating thermal reversibility of enantiotropic polymorphic pairs.
`Solubility
`of monotropic pairs as a function of temperature. The Ts is above melting point of both forms.
`Forms 1 and II are stable in entire temperature range and their corresponding melting points
`are shown. (d) On heating in DSC, form 11 melts (Tm,II) followed by recrystallization (Tcrys)
`and subsequent melting of form I (Tm,I). In cooling cycle only melting of form I is seen as
`an exotherm and on reheating only one endotherm corresponding to form I is seen. This is
`typical of monotropic polymorphs which converts from form 11 to stable form I and not vice
`versa.
`
`versible nature of these polymorphs. The heating rate in DSC is critical for char-
`acterizing the polymorphs, as a faster heating rate may not be able to identify the
`transition temperature, while a lower heating rate may lead to lower resolution of
`peaks. Therefore, it is a usual practice to generate DSC thermograms under differ-
`ent heating rates during polymorph characterization [11]. Also it is important to
`note that the sample preparation, particle size, and crucible type can affect the
`quality of the thermogram
`XRD is also another indispensable tool in identify-
`ing polymorphs. This is based on the differential scattering of X rays when passed
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 7 of 43)
`
`

`
`940
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`100
`
`Intensity
`
`01O
`
`100
`
`1 O
`
`20
`
`30
`
`30
`
`40
`
`50
`
`2 e
`
`(a)
`
`Intensity
`
`01O
`
`10
`
`20
`
`30
`
`30
`
`40
`
`50
`
`2 e
`
`(b)
`
`FIGURE 4 Schematic X—ray diffractograms of two polymorphic forms of a hypothetical
`drug. The x axis represents the detection angle and the y axis represents the intensity of the
`peak. As can be seen, there is a difference in the diffractograms due to the difference in the
`internal crystal lattice of polymorphs. The different internal arrangement in a crystal deflects
`the X ray at different angles.
`
`through a powder sample. Typically, on passing through a powder sample, X rays
`will tend to get diffracted at Various angles, and at some angle of detection, the X
`rays diffracted from the different planes of the crystal converge to form an ampli-
`fied signal, which is detected by a photomultiplier tube. The angles at which the
`XRD peaks are obtained are characteristic for a polymorph (Figures 4a & b). The
`sample should be uniformly spread to get a good X—ray diffractogram, as an
`improper sample preparation may lead to Variation in intensities due to the pre-
`ferred orientation of a crystal in the XRD sample holder [14]. Other techniques,
`such as infrared (IR) spectroscopy and solid—state nuclear magnetic resonauce
`(NMR), are also used to characterize the polymorphs and are listed in Table 2.
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 8 of 43)
`
`

`
`PHYSICAL/BULK CHARACTERISTICS
`
`941
`
`TABLE 2 Techniques to Characterize Different Crystalline Forms
`
`Technique
`
`Applications
`
`Thermal analysis
`Differential scanning Melting point, enthalpy of fusion, and crystallization; solid—state
`calorimetry
`transformations
`Thermogravimetric
`Stoichiometry of solvates and hydrates; identifying vaporization
`analysis
`and volatilization
`Visualization of solid—state transformations and desolvation
`Hot—stage microscopy
`events
`
`X—ray diffractometry
`
`Identifying polymorphs; quantification of degree of crystallinity;
`crystal lattice geometry and solid—state transformations
`
`Spectroscopy
`Infrared
`
`Near infrared
`
`Nuclear magnetic
`resonance
`
`Characterization of polymorphs based on functional groups;
`characterization of hydrates and solvates
`In situ analysis of solid—state conversions; identification and
`quantification of polymorphs in dosage forms
`Useful to understand difference in molecular arrangement of
`polymorphs, hydrates, and solvates
`
`TABLE 3 Difference in Solubilities of Polymorphs
`
`Drug
`
`Melting Point (°C)
`
`Solubility Ratio“
`
`Indomethacin
`Sulfathiazole
`Piroxicam
`
`157, 163
`177, 202
`136, 154
`
`1.1
`1.7
`1.3
`
`"Indicates ratio of solubility of low-melting polymorphic form to solu-
`bility of high-melting polymorphic form of drug.
`
`Polymorphism has significant implications in the solubility, bioavailability stabil-
`ity, processing, packaging, and storage of solid drug substances [15—17]. The met-
`stable polymorphic form may be used to improve the solubility of drug substances.
`Many drugs are known to exhibit polymorphism, particularly, steroids, barbiturates,
`anti—inflammatory drugs, and sulfonamides, which have a high probability of exhibit-
`ing polymorphism [15]. The existing knowledge on drug polymorphism is a good
`starting point for a preformulation scientist to anticipate polymorphs based on the
`drug chemistry. In some cases, polymorphism may provide an opportunity to improve
`the solubility of a drug. For example, form II of chloramphenicol palmitate has a
`higher dissolution rate resulting in significantly higher plasma concentration than
`form I when administered orally [15]. However, in many cases [16] the difference
`in solubility may not be significant enough to cause differences in oral bioavailability
`(Table 3). Although the polymorphs differ in their dissolution rates, it should be
`realized that once the drug goes into solution, they do not differ in their properties.
`If a drug’s absorption is limited by its poor membrane permeability, then the differ-
`ence in solubility of polymorphs may not impact its bioavailability. Similarly, if the
`drug dissolution is rapid in comparison to the gastrointestinal (GI) transit time, then
`the difference in polymorph solubility will not influence its bioavailability [16].
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 9 of 43)
`
`

`
`942
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`More than the presence of metastable polymorph, it is the conversion of the
`metastable to the stable form during processing, storage, or use that is of great
`concern to the pharmaceutical scientist [17]. The unpredictability in “conditions”
`that result in the generation and conversion of one polymorphic form to another
`mainly aggravates such a situation [18]. This is exemplified by ritonavir, which is a
`classical case of appearance of a “new polymorphic form” after the drug was mar-
`keted [19]. Ritonavir, an anti—retroviral, drug was introduced in the market as a
`polymorphic form I in soft gelatin capsule in 1996. Two years later, a new polymor-
`phic form II appeared in the formulation due to some unknown reasons causing the
`drug to be less soluble. The drug manufacturer withdrew the product due to failure
`of the batches in dissolution tests. After eXtensive investigation and reformulation,
`the drug was reintroduced in the market in 1999.
`Nonetheless, in spite of their unpredictability, a good preformulation team will
`be able to anticipate the different polymorphs during drug development. It should
`not be an impossible task given the recent advancements in high—throughput genera-
`tion and characterization of polymorphs [20]. From an innovator’s perspective, the
`identification and thorough characterization of multiple polymorphs during drug
`discovery can eXtend the patent life and delay the market entry of generic manu-
`facturers. On the other hand, it is also an opportunity for the generic manufacturers
`to generate new polymorphs with better solubility and stability for gaining market
`entry. The patent dispute on ranitidine polymorphs is a good eXample in this regard
`[21]. Two polymorphic forms of ranitidine were patented by the innovator company
`and the generic manufacturers had to find an appropriate method to manufacture
`the desired polymorph without the accompanying impurity of the other polymorph.
`This provided an edge for the innovator to eXtend the drug’s market eXclusivity for
`a little longer than they would otherwise have had. If a pure polymorph cannot be
`generated, the eXtent of polymorphic impurity should be quantified and ensured
`from batch to batch. The preformulation scientist closely works with the synthetic
`chemist in setting specifications for polymorphs.
`
`6.2.2.2 Hydrates/Solvates
`
`In addition to drug molecules, solvent molecules also get incorporated in the crystal
`lattice, resulting in altered physicochemical properties. When the solvent is water,
`they are known as hydrates, while if it is any other solvent, they are known as
`solvates. They are also known as pseudopolymorphs or solvatomorphs. Hydrates
`are important in this regard as one—third of all marketed drugs are hydrates [13].
`Depending on how the water is arranged inside the crystals, they are classified as
`isolated hydrates, channel hydrates, and ion—associated hydrates [13]. In isolated
`hydrates, the water molecules are separated from each other by the intervening drug
`molecules in the crystal lattice (e.g., cephadrine dihydrate). Channel hydrates result
`when water molecules are linked to one another forming a channel (e.g., theophyl—
`line monohydrate). The water molecules may be present either stoichometrically or
`nonstoichometrically within the crystal lattice. Ion—associated hydrates are typically
`seen when the water is metal ion coordinated (e.g., nedocromil zinc). Nonstoicho—
`metric channel hydrates are problematic due to the presence of diffusible water,
`which can easily move in and out of the crystal lattices [13, 22].
`Hydrates or solvates are formed by crystallizing the drug in the presence of water
`or solvents. The hydrate formation is dictated by water activity in a given solvent
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 10 of 43)
`
`

`
`PHYSICAL/BULK CHARACTERISTICS
`
`943
`
`
`
`
`
`HeatflowPercentweightloss
`
`Temperature (°C)
`
`FIGURE 5 Characterization of hydrate. (a) TGA thermogram of monohydrate. The ther-
`mogram shows weight loss as a function of temperature. The step in the thermogram shows
`weight loss due to dehydration of a hydrate. (b) DSC thermogram showing endotherm at
`corresponding temperature (Tdehyd). The second endotherm indicates the melting point of
`the hydrate (Tm).
`
`[22]. Hydrates are characterized using gravimetric methods such as thermogravi—
`metric analysis (TGA) or by Karl Fischer titrometry [23]. In TGA, the loss of
`water/solvate on heating a sample is recorded as a thermogram (Figure 5). The mass
`change due to dehydration is seen as a step loss in the TGA thermogram. Based on
`the weight of the initial sample and its elemental composition, the number of water
`molecules can be calculated. The TGA curve in combination with a DSC thermo-
`
`gram helps to differentiate hydrates from other thermal transitions. In Figure 5,
`the endotherm in the DSC thermogram corresponds to water loss as indicated
`by the TGA curve. The TGA can be coupled to an IR or mass spectrometer
`to characterize solvates. Thermal microscopy is a useful qualitative tool
`to
`visualize the release of water from the drug crystals as a function of temperature
`[23].
`Hydrate formation and dehydration significantly influences the processing and
`storage of drug products [17]. Hydrates may take up further water or dehydrate to
`lose water. Dehydration of hydrates leads to several possibilities [24], as shown in
`Figure 6. Hydrates on dehydration can form isomorphic desolvates retaining the
`same crystal lattice as the hydrate but without the water. Alternatively, hydrates can
`lose water and become anhydrous crystals. They can also lose water, forming amor-
`phates with the loss of crystal lattice. Higher hydrates can lose water to form lower
`hydrates, for example, pentahydrate converting to di— or monohydrate. The hydrates
`also can exhibit polymorphism or on dehydration can convert to a different poly-
`morphic form [13, 17]. Such solid—state transformations are possible during process-
`ing, such as granulation, tableting, and storage [17]. In general, hydrates are less
`soluble than anhydrous forms while solvates are more soluble than ansolvates in
`water. Ampicillin trihydrate is a classical example which shows lower solubility and
`lower plasma concentration than anhydrous ampicillin [15]. Preformulation studies
`
`Janssen Ex. 2044
`
`Lupin Ltd. v. Janssen Sciences Ireland UC
`|PR2015-01030
`
`(Page 11 of 43)
`
`

`
`944
`
`ROLE OF PREFORMULATION IN DEVELOPMENT OF SOLID DOSAGE FORMS
`
`Polymorphs
`
`Isomorphic
`
`desolvate v\
`
`Anhydrous
`
`/'
`
`Hydrate
`
`/ \
`
`Lower hydrate
`
`Amorphous
`
`FIGURE 6 Various possibilities that arise from dehydration of hydrate. A hydrate can
`dehydrate reversibly into various solid—state forms. It can dehydrate to form an anhydrous
`form of the drug or to a lower hydrate. Hydrate can also dehydrate to form an isomorphic
`desolvate where the crystal lattice is retained except for the absence of water. The crystal
`structure may also collapse on dehydration to form an amorphous form. Hydrates on dehy-
`dration can also result in different polymorphs.
`
`provide valuable inputs to the formulator in selecting a suitable form of the drug.
`For example, ampicillin is hygroscopic and hence can be used in suspension dosage
`forms, while ampicillin trihydrate, which is non—hygroscopic, is used in solid dosage
`forms.
`
`6.2.2.3 Amorphate
`
`Unlike a crystalline drug, an amorphous form of the drug does not have a regular
`crystal lattice arrangement and the molecules are arranged in random order (Figure
`1c). Glass is a typical amorphous substance and so amorphous drugs are also known
`as glasses [25]. Amorphous form is prepared through milling, rapid cooling of a melt,
`rapid precipitation using an antisolvent, rapid dehydration of a hydrate, spray drying,
`or freeze drying [26]. Some of the above methods may also unintentionally produce
`an amorphous form during processing of the crystalline form of the drug [17]. For
`instance, milling during dosage form manufacture may produce an amorphous form
`unintentionally, as in the case of indomethacin. The amorphous form does not show
`a melting point but is characterized by a glass transition temperature (Tg). This
`temperature indicates the conversion of the amorphous form from a rigid glassy
`state to a more mobile rubbery state. Above Tg the amorphous form will tend to
`recrystallize and convert to the crystalline form, which then undergoes melting, as
`shown in Figure 7a. The Tg for an amorphous drug can vary depending on the
`storage conditions and thermal history of the sample and is sensitive to moisture,
`pressure, and temperature [26]. The Tg is seen only as a slight shift in the baseline
`due to a change in the specific heat capacity of the sample and is influenced by the
`heating rate in DSC [25]. In XRD, the amorphous form shows a shallow peak
`or halo, as opposed to sharp and intense peaks for a crystalline drug compound
`(Figure 7b).
`The main advantage of amorphous form of the drug substance state is its signifi-
`cantly higher solubility than the crystalline form of the drug, primarily due to the
`excess surface energy [16, 27]. Therefore,

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