throbber
1F
`
`orm Selection of Pharmaceutical
`Compounds
`
`Ann W. Newman and G. Patrick Stahly
`SSCI, Inc., West Lafayette, Indiana
`
`I.
`
`INTRODUCTION
`
`The drug development process involves a number of activities which are carried
`out simultaneously, as shown by the oversimplified depiction in Fig. 1. Once a
`molecule is discovered that has desirable biological activity, the process of creat-
`ing a pharmaceutical drug product from this molecule begins. As toxicology and
`efficacy studies are undertaken, methods for manufacture of the active molecule
`and for its delivery in therapeutic doses are sought. Critical to the latter effort is
`finding a form of the active molecule which exhibits appropriate physical proper-
`ties. The form ultimately selected, called the active pharmaceutical ingredient
`(API), or drug substance, must be stable and bioavailable enough to be formulated
`into a drug product, such as a tablet or suspension. This formulation must be
`effective at delivering the active molecule to the targeted biosystem.
`This chapter describes methodology useful in selection of the appropriate
`solid form of a drug substance for inclusion in a drug product. Form selection
`is commonly considered among the primary goals of a preformulation study.
`However, the investigative techniques discussed herein also have application in
`early drug substance and drug product development activities (shown by the cir-
`cled area in Fig. 1).
`Solid form selection involves the preparation and property evaluation of
`many derivatives of an active molecule. Drug substance properties of importance
`in the drug development process may be categorized as shown in Table 1. These
`properties depend on the nature of the drug substance and the final formulation.
`Many bioactive organic molecules contain ionizable groups such as carboxylic
`
`Merck Exhibit 2218, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 1 The drug development process.
`
`acid or amino groups. Reaction of these compounds with acids or bases produce
`salts, which have much different physical properties than the neutral parents. A
`single molecular entity, be it a salt or a neutral molecule, often exists in multiple
`solid forms, each of which exhibits unique physical properties. The properties
`of many such forms need to be evaluated relative to the intended formulation.
`A lyophilized product that will be dissolved and injected needs to be chemically
`stable in the dry state and adequately soluble in the carrier. On the other hand,
`the drug substance in a tablet formulation needs to be processable, chemically
`stable, and physically stable in the dry state, as well as having adequate solubility
`for delivery.
`Form selection activities should be started as early in the development pro-
`cess as material availability allows. Salt selection, including preparation and eval-
`
`Table 1 Some Important Properties of Drug Substances
`
`Bioavailability
`
`Chemical and physical stability
`
`Processibility
`
`Dissolution rate
`Solubility
`Toxicity
`
`Excipient compatibility
`Hygroscopicity
`Oxidative stability
`Photostability
`Thermodynamic stability
`Crystal form
`
`Color
`Compactibility
`Density
`Ease of drying
`Filterability
`Flowability
`Hardness
`Melting point
`Particle size
`
`Merck Exhibit 2218, Page 2
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`uation of samples, and polymorph screening can be carried out with as little as
`half a gram of active compound. Results of form selection include information
`that can be used in planning the final step of the manufacturing process (often
`crystallization) as well as information that is critical to formulation development.
`The nature and extent of work to be performed during development can
`be modeled after the draft International Committee on Harmonization (ICH) Q6A
`document on specifications, which can be found on the Food and Drug Adminis-
`tration (FDA) website (www.fda.cder.gov). This document outlines the specifi-
`cations needed for a New Drug Application and contains several decision trees
`to guide the selection of specifications. The Q6A decision tree 4 (Fig. 2) describes
`
`Fig. 2 Flow chart 4 from the ICH Q6A document (www.fda.cder.gov).
`
`Merck Exhibit 2218, Page 3
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 2 Continued
`
`methods for the study of solids for a polymorph screen as well as characterization
`of the drug substance in the drug product. Other decision trees have also been
`reported in the literature (1).
`In this chapter we describe the form selection process. A short review of
`the analytical techniques commonly employed is followed by sections covering
`salt and solid form selection. Form selection should be approached in a planned,
`rational manner, but it is important to realize that not all compounds will allow
`adherence to a single experimental plan. The exercise is a scientific one, and it
`will yield the best results only if carried out with judgment and flexibility.
`
`II. ANALYTICAL TECHNIQUES
`
`A number of analytical techniques are commonly used in form selection studies.
`Various publications (2–4) and books (5,6) describe physical characterization
`
`Merck Exhibit 2218, Page 4
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`of solid-state pharmaceuticals. A brief description of common methods will be
`presented in this section.
`
`A. X-Ray Diffraction
`
`Crystalline organic solids are made up of molecules which are packed or ordered
`in a specific arrangement. These molecules are held together by relatively weak
`forces, such as hydrogen bonding and van der Waals interactions. The arrange-
`ment of the molecules is defined by a unit cell, which is the smallest repeating
`unit of a crystal. The unit cell can be divided into planes, as shown in Fig. 3.
`X-ray diffraction techniques used for characterizing pharmaceutical solids
`
`Fig. 3 A packing diagram of unit cells divided into planes.
`
`Merck Exhibit 2218, Page 5
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`(1)
`
`(n ⫽ 1, 2, 3, . . .)
`
`include the analysis of single crystals and powders. The electrons surrounding
`the atoms diffract X-rays in a manner described by the Bragg equation:
`nλ ⫽ 2d sin θ
`where
`λ ⫽ X-ray wavelength
`d ⫽ spacing between the diffracting planes
`θ ⫽ diffraction angle
`A schematic of the diffraction phenomenon is given in Fig. 4. X-rays will be
`diffracted at an angle defined as θ. Knowing the diffraction angle and the X-ray
`wavelength, the spacing between the planes can be calculated. Conditions of the
`Bragg equation must be satisfied to achieve constructive interference of the dif-
`fracted X-rays and produce a beam that can be measured by the detector. If the
`conditions of the Bragg equation are not satisfied, diffracted waves interfere de-
`structively, with a net diffracted intensity of zero.
`For single-crystal diffraction, a good-quality single crystal of the sample
`of interest is required. From the angles and intensities of diffracted radiation, the
`structure of the crystal can be elucidated and the positions of the molecules in
`the unit cell can be determined. The result is often displayed graphically as the
`asymmetric unit, which is the smallest part of a crystal structure from which
`the complete structure can be obtained using space-group symmetry operations.
`
`Fig. 4 A schematic representation of X-ray diffraction.
`
`Merck Exhibit 2218, Page 6
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`The unit cell parameters, the lengths (a, b, c) as well as the angles (α, β, γ) of
`the unit cell are also determined from the crystal structure. There are seven classes
`of unit cells: triclinic, monoclinic, orthorhomic, tetragonal, hexagonal, rhombo-
`hedral, and cubic. For pharmaceutics, only triclinic (a ≠ b ≠ c, α ≠ β ≠ γ ≠90°),
`monoclinic (a ≠ b ≠ c, α ≠ γ ≠90°, β ⫽ 90°), and orthorhombic (a ≠ b ≠ c,
`α ⫽ β ⫽ γ ⫽ 90°) unit cells are commonly observed.
`The unit cells can be ‘‘packed’’ into a three-dimensional display of the
`crystal lattice. The orientation of the molecules is responsible for various proper-
`ties of the crystalline substance. For example, hydrogen bonding networks may
`provide high stability, and spaces in the structure may allow easy access of small
`molecules to provide hydrated or solvated forms.
`Crystal structures provide important and useful information about solid-
`state pharmaceutical materials. Unfortunately, it is not always possible to grow
`suitable single crystals of a drug substance. In these cases, X-ray diffraction of
`powder samples can be used for comparison of samples.
`X-ray powder diffraction (XRPD) is the analysis of a powder sample. The
`typical output is a plot of intensity versus the diffraction angle (2θ). Such a plot
`can be considered a fingerprint of the crystal structure, and is useful for determi-
`nation of crystallographic sameness of samples by pattern comparison. A crystal-
`line material will exhibit peaks indicative of reflections from specific atomic
`planes. The patterns are representative of the structure, but do not give positional
`information about the atoms in the molecule. One peak will be exhibited for all
`repeating planes with the same spacing. An amorphous sample, on the other hand,
`will exhibit a broad hump in the pattern called an amorphous halo, as shown in
`Fig. 5.
`
`Fig. 5 The XRPD pattern exhibited by an amorphous material.
`
`Merck Exhibit 2218, Page 7
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`XRPD is dependent on a random orientation of the particles during analysis
`to obtain a representative powder pattern. The sample, as well as sample prepara-
`tion, can greatly effect the resulting pattern. Large particles or certain particle
`morphologies, such as needles or plates, can result in preferred orientation. Pre-
`ferred orientation is the tendency of crystals to pack against each other with some
`degree of order and it can affect relative peak intensities, but not peak positions,
`in XRPD patterns. If a powder is packed into an XRPD sample holder and the
`surface is smoothed with a microscope slide or similar device, crystals at the
`surface can become aligned so that a nonstatistical arrangement of crystal faces
`is presented to the X-ray beam. The result is that some reflections are artificially
`intensified and others are artificially weakened. One way to determine if preferred
`orientation is causing relative peak intensity changes is to grind and reanalyze
`samples. Grinding reduces particle size and disrupts the crystal habit, both of
`which tend to minimize preferred orientation effects. However, grinding can
`cause crystal form changes, so care must be taken to interpret the patterns with
`this in mind. The effects of preferred orientation can be profound, as illustrated
`by the XRPD patterns shown in Fig. 6.
`
`Fig. 6 XRPD patterns of the same sample before (top) and after (bottom) grinding. The
`polymorphic form of the sample was not changed by grinding.
`
`Merck Exhibit 2218, Page 8
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Qualitative analysis of powder patterns can be used to determine if multiple
`samples are the same crystal form or if multiple crystal forms have been pro-
`duced. Mixtures of samples can also be evaluated. When mixtures are obtained,
`XRPD can also be used in a quantitative mode to calculate the amount of each
`phase present.
`
`B. Thermal Methods
`
`Thermal methods of analysis discussed in this section are differential scanning
`calorimetry (DSC), thermogravimetry (TG), and hot-stage microscopy (HSM).
`All three methods provide information upon heating the sample. Heating can be
`static or dynamic in nature, depending on the information required.
`Differential scanning calorimetry monitors the energy required to maintain
`the sample and a reference at the same temperature as they are heated. A plot
`of heat flow (W/g or J/g) versus temperature is obtained. A thermal transition
`which absorbs heat (melting, volatilization) is called endothermic. If heat is re-
`leased during a thermal transition (crystallization, degradation), it is called exo-
`thermic. The area under a DSC peak is directly proportional to the heat absorbed
`or released and integration of the peak results in the heat of transition.
`Samples are loaded into pans for DSC analysis. Pan configuration (open,
`crimped, hermetically sealed, hermetically sealed with a pinhole, etc.) and scan
`rate can result in variations in position and intensity of the thermal events. These
`variations can be used to gain further information about the sample as well as
`other crystal forms.
`The observance of thermal transitions by DSC is insufficient to fully char-
`acterize the behavior of a substance on heating. It is not known if an endothermic
`transition observed in the DSC is a volatilization or a melt without corroborating
`information, such as TG or HSM data. It is important to understand the origin
`of the DSC transitions to fully characterize the system and understand the rela-
`tionship between various solid forms.
`Thermogravimetry measures the weight change of a sample as a function
`of temperature. A total volatile content of the sample is obtained, but no informa-
`tion on the identity of the evolved gas is provided. The evolved gas must be
`identified by other methods, such as gas chromatography, Karl Fisher titration
`(specifically to measure water), TG–mass spectroscopy, or TG–infrared spectros-
`copy. The temperature of the volatilization and the presence of steps in the TG
`curve can provide information on how tightly water or solvent is held in the
`lattice. If the temperature of the TG volatilization is similar to an endothermic
`peak in the DSC, the DSC peak is likely due or partially due to volatilization.
`It is usually necessary to utilize multiple techniques to determine if more than
`one thermal event is responsible for a given DSC peak.
`
`Merck Exhibit 2218, Page 9
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Hot-stage microscopy is a technique that supplements DSC and TG. Events
`observed by DSC and/or TG can be readily characterized by HSM. Melting, gas
`evolution, and solid–solid transformations can be visualized, providing the most
`straightforward means of identifying thermal events. Many polymorphic systems
`have been investigated using only these thermal methods, as illustrated by the
`publications of Kuhnert-Brandsta¨tter (7). Details of the methodologies used in
`hot-stage microscopy have also been reviewed (8).
`Thermal analysis can be used to determine the melting points, recrystal-
`lizations, solid-state transformations, decompositions, and volatile contents of
`pharmaceutical materials. DSC can also be used to analyze mixtures quantita-
`tively.
`
`C. Vibrational Spectroscopy
`
`Common methods used to characterize drugs and excipients are infrared (IR) and
`Raman spectroscopy. These techniques are sensitive to the structure, conforma-
`tion, and environment of organic compounds. Because of this sensitivity, they
`are useful characterization tools for pharmaceutical crystal forms. Qualitative as
`well as quantitative analysis can be performed with both techniques.
`Infrared spectroscopy is based on the conversion of IR radiation into molec-
`ular vibrations. For a vibration to be IR-active, it must involve a changing molec-
`ular dipole (asymmetric mode). For example, vibration of a dipolar carbonyl
`group is detectable by IR spectroscopy. Whereas IR has been traditionally used
`as an aid in structure elucidation, vibrational changes also serve as probes of
`intermolecular interactions in solid materials.
`Sampling techniques for IR include pellets, mulls, and diffuse reflectance.
`Diffuse reflectance is the best choice for crystal form determination, due to the
`minimal sample manipulation required. Mulls can also be used for form identifi-
`cation, but peaks due to the suspension medium may interfere with the peaks of
`interest.
`Raman spectroscopy is based on the inelastic scattering of laser radia-
`tion with loss of vibrational energy by a sample. A vibrational mode is Raman-
`active when there is a change in the polarizability during the vibration. Symmetric
`modes tend to be Raman-active. For example, vibrations about bonds be-
`tween the same atom, such as in alkynes, can be observed by Raman spectros-
`copy.
`
`Small amounts of samples can be analyzed by Raman spectroscopy and a
`variety of sample holders are available, ranging from stainless steel holders to
`glass NMR tubes. The samples are analyzed neat, eliminating the need for sample
`preparation procedures that may induce solid form changes. Since a laser is used,
`only a small portion of the sample is in the beam during analysis.
`
`Merck Exhibit 2218, Page 10
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`D. Nuclear Magnetic Resonance (NMR) Spectroscopy
`
`NMR spectroscopy probes atomic environments based on the different resonance
`frequencies exhibited by nuclei in a strong magnetic field. Many different nuclei
`are observable by the NMR technique, but those of hydrogen and carbon atoms
`are most frequently studied. NMR spectroscopy of solutions is commonly used
`for structure elucidation. However, solid-state NMR measurements are extremely
`useful for characterizing the crystal forms of pharmaceutical solids.
`Nuclei that are typically analyzed with this technique include those of 13C,
`31P, 15N, 25Mg, and 23Na. Different crystal structures of a compound can result in
`perturbation of the chemical environment of each nucleus, resulting in a unique
`spectrum for each form. Once resonances have been assigned to specific atoms
`of the molecule, information on the nature of the polymorphic variations can be
`obtained. This can be useful early in drug development, when the single-crystal
`structure may not be available. Long data acquisition times are common with
`solid-state NMR, so it is often not considered for routine analysis of samples.
`However, it is usually a very sensitive technique, and sample preparation is mini-
`mal. NMR spectroscopy can be used either qualitatively or quantitatively, and
`can provide structural data, such as the identity of solvents bound in a crystal.
`
`E. Moisture Sorption/Desorption and Hygroscopicity
`
`Hygroscopicity and the formation of hydrated crystal forms can be investigated
`by means of moisture sorption/desorption methods. Sample analysis may be car-
`ried out using automated equipment or by periodic weighing of samples kept
`over saturated salt solutions providing various relative humidities (RHs). In either
`case, water taken in or released by a sample is detected as a change in sample
`weight. If a material readily loses water of hydration at low relative humidity,
`the stability of the hydrate may need to be investigated further. If a material
`readily gains moisture at ambient or high relative humidity, hygroscopicity stud-
`ies will be needed to determine if a change in crystal form is associated with the
`water uptake. This is done by characterizing material equilibrated under various
`relative humidity conditions using techniques suitable for detection of crystal
`form, such as XRPD, TG, DSC, and IR spectroscopy. Changes in water content
`and crystal form may lead to definition of specific handling conditions under
`which a change in form will not occur.
`
`F. Summary
`
`Only a brief description of selected techniques for solid-state characterization
`has been given above. Many other techniques are available. It is imperative that
`
`Merck Exhibit 2218, Page 11
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`a multidisciplinary approach be applied to the characterization solids; no single
`analytical method can provide all the information necessary to understand the
`nature and properties of solid pharmaceutical compounds.
`
`III. SALT SELECTION
`
`A. Factors Guiding Salt Selection
`
`Salt selection is a critical part of the drug development process because selection
`of an appropriate salt can significantly reduce time to market. Changing salts in
`the middle of a development program may require repeating most of the biologi-
`cal, toxicological, formulation, and stability studies performed initially. However,
`continuing the development of a nonoptimal salt may lead to increased develop-
`mental and production costs, even product failure. Selection of the correct salt
`early in the development process will avoid these problems and facilitate down-
`stream development activities. In addition, salts that exhibit advantageous proper-
`ties are usually patentable as new chemical entities.
`Salts are used to alter the physical, chemical, biological, and economic
`properties of a drug substance. The change in crystal structure accomplished by
`forming a salt can lead to greatly improved properties. The advantages of using
`salt forms in pharmaceutical formulations have been extensively reviewed (9).
`A variety of factors can guide the salt selection process and a partial list of consid-
`erations is given in Table 2.
`A change in the solubility of a drug substance is often a major reason for
`choosing a salt. In many cases, substances containing free acid or base groups
`have poor aqueous solubility which saltification of these groups can improve,
`leading ultimately to greater bioavailability. Increasing the solubility of a weak
`acid–base drug substance by forming a variety of salts has been reported for
`
`Table 2 Factors Guiding the Salt Selection
`Process
`
`Bioavailability
`Chemical stability
`Crystallinity
`Dissolution rate
`Cost
`Handling properties
`Hygroscopicity
`Melting point
`Intended formulation
`
`pH of salt solutions
`Physical stability
`Processing properties
`Purity
`Solubility
`Taste
`Toxicity
`Wettability
`Yield
`
`Merck Exhibit 2218, Page 12
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 7 The structure of RS-82856.
`
`RS-82856 (Fig. 7) (10). Five salts (chloride, hydrogen sulfate, phosphate, sodium,
`and potassium) exhibited significantly higher solubility and dissolution rates than
`the parent drug. Based on a variety of physical parameters (solubility, dissolution
`rate, melting point, hygroscopicity, and chemical stability), the hydrogen sulfate
`form was recommended for development. A bioavailability study in dogs compar-
`ing the parent drug and the hydrogen sulfate salt resulted in the salt being ab-
`sorbed approximately two to three times more efficiently than the parent drug.
`The solubility and dissolution data were good indicators of the bioavailability of
`this material.
`For some drugs, preparation of stable salts may not be feasible, or free acid
`or free base forms may be preferred. A reported example compares the free base
`and hydrochloride salt of the poorly water-soluble drug, α-pentyl-3-(2-quino-
`linylmethoxy)benzenemethanol, known as REV 5901 (Fig. 8) (11). For this drug
`substance, lower solubility of the chloride salt, along with equivalent dissolution
`rates, resulted in the free base being chosen for development.
`It should be noted that a salt usually exhibits a higher melting point than
`the free acid or base, which can result in greater stability and easier processing.
`However, there is often a relationship between melting point and aqueous solubil-
`ity. Gould, in his study of the salts of basic drugs, concluded that ‘‘ideal solubility
`of a drug in all solvents decreases by an order of magnitude with an increase of
`100°C in its melting point’’ (12). An example of this phenomenon is the antima-
`larial drug α-(2-piperidyl)-3,6-bis(trifluoromethyl)-9-phenanthrenemethanol hy-
`
`Fig. 8 The structure of REV 5901.
`
`Merck Exhibit 2218, Page 13
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 9 The
`methanol.
`
`structure of α-(2-piperidyl)-3,6-bis(trifluoromethyl)-9-phenanthrene-
`
`drochloride (Fig. 9). The melting point and solubility data are shown in Table 3
`(13). Overall, a substantial decrease in solubility was observed with the increase
`in melting point of the salts. It should also be noted that the solubility of salts
`can be affected not only by changing the lattice energy (melting point), but also
`by enhancing water–drug interactions. The study of chlorhexidine (Fig. 10)
`showed that the solubility of this drug was significantly enhanced by increasing
`the number of hydroxyl groups on the conjugate acid (14).
`The melting point of a drug substance salt can be greatly influenced by the
`counterion. For UK47880 (Fig. 11), a relationship was observed between the
`melting points of the salts and the corresponding conjugate acid (12). Salts pre-
`
`Table 3 Melting-Point and Solubility Data for
`α-(2-piperidyl)-3,6-bis(trifluoromethyl)-9-phenanthrenemethanol
`hydrochloride (13)
`
`Salt form
`
`Free base
`DL-Lactate
`L-Lactate
`2-Hydroxyethane sulfonate
`Sulfate
`Mesylate
`Hydrochloride
`
`Melting point
`of salt (°C)
`
`215
`172
`192
`251
`270
`290
`331
`
`Aqueous
`solubility
`(mg/mL)
`
`7.5
`1850
`925
`620
`20
`300
`13
`
`Merck Exhibit 2218, Page 14
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 10 The structure of chlorhexidine.
`
`Fig. 11 The structure of UK47880.
`
`pared from high-melting aromatic acids exhibited higher melting points, whereas
`salts prepared from low-melting flexible aliphatic acids yielded oils. In the case
`of epinephrine (Fig. 12), the effect of hydrogen bonding on the melting points
`of the salts was apparent (12). Small acids prone to form hydrogen bonds (ma-
`lonic and maleic) resulted in higher-melting salts. The bitartrate and fumarate
`salts were found to be lower-melting due to their size and possibly unfavorable
`symmetry, respectively.
`A salt can also provide improved chemical stability compared to the parent
`drug substance. An example of this was reported for xilobam, whose structure
`is shown in Fig. 13 (15). In order to protect xilobam from the effects of high
`temperature and humidities without decreasing the dissolution rate, three arylsul-
`fonic acid salts (tosylate, 1-napsylate, and 2-napsylate), as well as the saccharate
`salt, were prepared. The 1-napsylate was found to be the most chemically stable
`form at 70°C and 74% RH after 7 days. Dissolution data from compressed tablets
`
`Fig. 12 The structure of epinephrine.
`
`Merck Exhibit 2218, Page 15
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 13 The structure of xilobam.
`
`showed that the 1-napsylate salt released xilobam at a faster rate than the free
`base. This work demonstrated that a strong acid with an aryl group protected the
`easily hydrolyzed base from the effects of high temperature and humidity.
`A choice of salts can also expand the formulation options for a material.
`The antimalarial agent α-(2-piperidyl)-3,6-bis(trifluoromethyl)-9-phenanthrene-
`methanol hydrochloride (Fig. 9) exhibited poor solubility, was delivered as an
`oral formulation, and required a single dosing of 750 mg (13). Seven salts and
`the free base were evaluated. The lactate salt was found to be 200 times as soluble
`as the hydrochloride salt (Table 3). This enhanced solubility would make it possi-
`ble to reduce the oral dose to achieve the same therapeutic response as well as
`develop a parenteral formulation for the treatment of malaria. However, the case
`of lidocaine hydrochloride (Fig. 14) demonstrates that a compound limited to
`parenteral and topical formulations can be expanded to oral administration by
`changing to a salt form with acceptable physical properties (16). The hydrochlo-
`ride salt was hygroscopic, difficult to prepare, and hard to handle. Six salts were
`evaluated for salt formation, solubility, and hygroscopicity. Other salts, such
`as phosphate, exhibited properties acceptable for dry pharmaceutical dosage
`forms.
`Many other examples can be found in the literature that demonstrate the
`applicability of examining a number of salts to obtain the necessary properties
`needed for development and marketing of the drug substance. Excellent reviews
`on salts (9,12) discuss many of the issues involved in targeting salt forms of drug
`substances.
`
`Fig. 14 The structure of lidocaine.
`
`Merck Exhibit 2218, Page 16
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`Merck Exhibit 2191, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`B. Counterions
`
`Salt formation involves proton transfer from an acid to a base. In theory, any
`compound that exhibits acidic or basic characteristics can form salts. The major
`consideration is the relative acidity and/or basicity of the chemical species in-
`volved. To form a salt, the pKa of the acidic partner must be less than the pKa
`of the conjugate acid of the basic partner. These pKa values need to be about
`two units apart for total proton transfer to occur, otherwise an equilibrium mixture
`of all components (acid, base, and salt) is likely to result. Even so, equilibrium
`mixtures of this type can often be used to prepare salts if a driving force is present,
`such as the crystallization of the salt from solution.
`Another consideration is the toxicity of the counterion. A large number of
`anions and cations are available for pharmaceutical compounds, and tabulations
`of those approved by the FDA have appeared in the literature (9,12). An expanded
`but not comprehensive list of acceptable ions is presented in Table 4. In general,
`ions related to normal metabolic chemicals or present in food or drink are usually
`regarded as suitable candidates for preparing salts.
`Target salts are chosen by considering a number of factors. The structure
`and pKa of the drug substance are important values to determine initially. Avail-
`able literature on structurally related compounds can result in excellent leads for
`target salts. The chemical stability of the drug substance, especially as related to
`pH stability, will also play a role. The ease of large-scale preparation of the salt,
`as well as the cost of the counterion and processing, will need to be considered
`to determine if the salt is a feasible choice. The type of drug product and antici-
`pated loading of the drug substance in the drug product can also influence the
`choice of salts. For high drug loadings, a large, bulky counterion, which adds
`substantial mass to the loading, may not be the best choice. Anions that irritate
`the gastrointestinal tract should be avoided for certain drugs, such as anti-
`inflammatories. The relative acid/base strength of the resulting salt and the ten-
`dency to disproportionate should be considered when using basic excipients in
`a formulation.
`A common salt choice for basic drug substances is the hydrochloride, be-
`cause of its availability. However, a number of issues also need to be considered
`when using this salt. Reports have shown that hydrochloride salts do not always
`increase the solubility of poorly soluble basic drugs (1,13,17,18), due to the com-
`mon-ion effect. The presence of chloride ions in the gastric fluid can result in a
`lower solubility for the hydrochloride salt.
`The hydrochloride salt is often a stronger acid than is needed for many
`drug substances, which can result in low pH values for the aqueous solutions.
`This can lead to limitations in parenteral formulations or processing. The highly
`polar nature of hydrochloride salts can also l

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