throbber
Advanced Drug Delivery Reviews 59 (2007) 603 – 616
`
`www.elsevier.com/locate/addr
`
`Salt formation to improve drug solubility ☆
`Abu T.M. Serajuddin ⁎
`
`Science, Technology and Outsourcing Section, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ 07936, USA
`
`Received 23 April 2007; accepted 10 May 2007
`Available online 29 May 2007
`
`Abstract
`
`Salt formation is the most common and effective method of increasing solubility and dissolution rates of acidic and basic drugs. In this article,
`physicochemical principles of salt solubility are presented, with special reference to the influence of pH–solubility profiles of acidic and basic
`drugs on salt formation and dissolution. Non-ideality of salt solubility due to self-association in solution is also discussed. Whether certain acidic
`or basic drugs would form salts and, if salts are formed, how easily they would dissociate back into their free acid or base forms depend on
`interrelationships of several factors, such as S0 (intrinsic solubility), pH, pKa, Ksp (solubility product) and pHmax (pH of maximum solubility). The
`interrelationships of these factors are elaborated and their influence on salt screening and the selection of optimal salt forms for development are
`discussed. Factors influencing salt dissolution under various pH conditions, and especially in reactive media and in presence of excess common
`ions, are discussed, with practical reference to the development of solid dosage forms.
`© 2007 Elsevier B.V. All rights reserved.
`
`Keywords: Salt; solubility; pH–solubility profile; Common-ion effect; Self-association; Dissolution rate; Salt selection; Counterion; Microenvironmental pH
`
`Contents
`
`1.
`2.
`
`3.
`
`4.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Principles of salt formation and salt solubility .
`pH–solubility interrelationship of free base and its salt .
`.
`.
`2.1.
`pH–solubility interrelationship of free acid and its salt .
`.
`.
`2.2.
`.
`.
`2.3.
`Effect of counterion on salt solubility .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Effects of solubility, pKa and Ksp on pHmax .
`.
`.
`.
`.
`.
`.
`2.4.
`2.5. Deviation of pH–solubility interrelationship from ideality .
`Structure–solubility relationships .
`2.6.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2.7.
`Effect of organic solvent on salt solubility .
`.
`.
`.
`.
`.
`.
`.
`.
`Principles of salt dissolution .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1. General solubility–dissolution rate relationships .
`.
`.
`.
`.
`.
`3.2. Dissolution in reactive media .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.
`Common-ion effect on dissolution of salts .
`.
`.
`.
`.
`.
`.
`.
`.
`Solubility considerations in salt screening .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.1.
`Identification of chemical form .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.2. Determination of salt solubility .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.3.
`Recent trends in salt forms .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`604
`604
`604
`605
`606
`606
`607
`608
`609
`609
`609
`610
`611
`611
`612
`613
`613
`
`☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Drug solubility: How to measure it, how to improve it”.
`⁎ Tel.: +1 862 778 3995; fax: +1 973 781 7329.
`E-mail address: abu.serajuddin@novartis.com.
`
`0169-409X/$ - see front matter © 2007 Elsevier B.V. All rights reserved.
`doi:10.1016/j.addr.2007.05.010
`
`Merck Exhibit 2193, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`604
`
`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`5.
`
`Practical considerations of salt solubility in dosage form design .
`5.1.
`Liquid formulations .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.2. Microenvironmental pH of salts in solid dosage forms .
`.
`6. Conclusions and outlook .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`. 614
`. 614
`. 614
`. 614
`. 615
`
`1. Introduction
`
`Salts of acidic and basic drugs have, in general, higher solu-
`bilities than their corresponding acid or base forms. Salt formation
`to increase aqueous solubility is the most preferred approach for
`the development of liquid formulations for parenteral adminis-
`tration [1]. For solid dosage forms, Nelson [2,3] demonstrated as
`early as in 1950s that dissolution rates of salt forms of several
`weakly acidic compounds under gastrointestinal (GI) pH con-
`ditions were much higher than those of their respective free acid
`forms. He attributed the higher dissolution rate of a salt to its
`higher solubility (relative to the free acid form) in the aqueous
`diffusion layer surrounding the solid. Pronounced differences
`were observed in rates and extents of absorption of novobiocin [4]
`and tolbutamide [5] as compared to their respective sodium salts.
`Monkhouse and coworkers [6,7] reviewed physicochemical and
`biopharmaceutical advantages of salts over their free acid or base
`forms. The interest in salt formation has grown greatly over the
`past half a century and, in recent years, it has become the most
`commonly applied technique of increasing solubility and dis-
`solution rate in drug product development.
`The primary reason for the increased interest in salt formation
`is that with the progress in medicinal chemistry and, especially
`due to the recent introduction of combinatorial chemistry and
`high-throughput screening in identifying new chemical entities
`(NCE) [8,9], the solubility of new drug molecules has decreased
`sharply [10]. While a value of less than 20 μg/mL for the solu-
`bility of a NCE was practically unheard of until the 1980s, the
`situation has changed so much that in the present day drug
`candidates with intrinsic solubilities (solubility of neutral or
`unionized form) of less than 1 μg/mL are very common [11].
`Lipinski [12] reported that 31.2% of a group of 2246 compounds
`synthesized in academic laboratories between 1987 and 1994 had
`solubility equal to or less than 20 μg/mL. According to the recent
`experience of the present author, approximately one-third of new
`compounds synthesized in medicinal chemistry laboratories have
`an aqueous solubility less than 10 μg/mL, another one-third have
`a solubility from 10 to 100 μg/mL, and the solubility of the
`remaining third is N100 μg/mL. With such a predominance of
`poorly water-soluble compounds, careful attention must be paid
`to identification and selection of optimal salt forms for de-
`velopment. In certain cases, salt formation may not be feasible due
`to physical and chemical properties of NCEs. In other cases, even
`though salts can be synthesized, they may not serve the purpose of
`enhancing dissolution rate and bioavailability. It is important that
`the reasons behind such situations are understood.
`Despite major advantages of the use of salts, only limited
`attention has been paid historically to the selection of optimal
`salt forms for pharmaceutical product development [6,13]. At
`
`the beginning of drug development programs, salts were often
`selected based on ease of synthesis, ease of crystallization, cost
`of raw material, etc., and no systematic studies to evaluate their
`physicochemical properties, such as physical and chemical
`stability, processability into dosage forms, solubility and dis-
`solution rate at different pH conditions, etc., were conducted. If
`a salt was later found to be suboptimal for the desired formu-
`lation or if problems developed, it was often difficult to change
`the salt form without delaying the drug development program,
`since it required repeating most of the biological, toxicological,
`formulation and stability tests that had already been performed
`[14]. For most practical purposes, identification and selection of
`salt forms of NCEs still remain a trial and error process.
`One major objective of the present article is to review the basic
`principles of salt formation and how salts influence solubility and
`dissolution rate in a comprehensive manner, such that they can be
`easily applied to the development of drug substances as well as
`dosage forms. Efforts will be made to indicate the application of
`such principles in screening various salt candidates for a NCE,
`identification of optimal salt form, and ultimately formulation of
`dosage forms using the selected salt. Wherever possible, advan-
`tages and disadvantages of salt forms relative to their respective
`free acid or base forms will be presented.
`One particular issue with the use of salts in drug development
`is that, while salts are usually prepared from organic solvents,
`they are destined to encounter aqueous environment (water,
`humidity) during dosage form development and, in case of an
`orally administered tablet or capsule, at the time of dissolution in
`GI fluid. Therefore, a perfectly good salt isolated from an or-
`ganic solvent may not behave well in an aqueous environment
`due to low solubility, conversion to free acid or base forms, poor
`stability, etc., thus limiting its use in dosage forms.
`
`2. Principles of salt formation and salt solubility
`
`The aqueous solubility of an acidic or basic drug as a function
`of pH dictates whether the compound will form suitable salts or
`not and, if salts are formed, what some of their physicochemical
`properties might be [15]. pH–solubility interrelationships also
`dictate what counterions would be necessary to form salts, how
`easily the salts may dissociate into their free acid or base forms,
`what their dissolution behavior would be under different GI pH
`conditions, and whether solubility and dissolution rate of salts
`would be influenced by common ions [15,16].
`
`2.1. pH–solubility interrelationship of free base and its salt
`
`Kramer and Flynn [17] demonstrated that the pH–solubility
`profile of a basic drug may be expressed by two independent
`
`Merck Exhibit 2193, Page 2
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Fig. 2. pH–solubility profiles of haloperidol determined by using methane-
`sulfonic (mesylic) (□), hydrochloric (○) and phosphoric (▵) acids (reproduced
`from Ref. [24] with permission).
`
`only one point, can both the free base and salt coexist as solids.
`If the pH of a saturated solution with excess solid free base is
`lowered from above the pHmax to below the pHmax, the solid
`phase will convert to the salt, and it is important to note here
`that the pH will not drop below pHmax until enough acid is
`added to convert the entire excess free solid base into salt. The
`reverse is true for the conversion of a salt to the free base; no
`free base will precipitate out until the pH is raised above the
`pHmax.
`There are numerous reports in the literature confirming
`interrelationships of solubilities of bases and their salt forms
`as per the schematics in Fig. 1 [17,18–23]. An example of
`typical pH–solubility profiles is given in Fig. 2, where solu-
`bilities of haloperidol and its methanesulfonate (mesylate),
`hydrochloride and phosphate salts as a function of pH are
`shown [24].
`
`2.2. pH–solubility interrelationship of free acid and its salt
`
`Fig. 3 shows a schematic diagram for the pH–solubility
`interrelationship of a free acid and its salt form. The free acid
`would be the equilibrium species at a pH below pHmax, and it
`would convert to a salt only if it is equilibrated with a solution at
`a pH above pHmax by adding a sufficient quantity of an alkali or
`
`where BH+ and B represent, respectively, protonated (salt) and
`free base forms of the compound. When the aqueous medium at
`a given pH is saturated with the free base, the total solubility
`(ST) at that pH may be expressed as follows:
`

`ST; base pH N pH max
`
`s
`
`
`s 1 þ H3O
`Ka
`
`
`

`
`ð3Þ
`
`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`605
`
`Fig. 1. Schematic representation of the pH–solubility profile of a basic drug
`indicating that the solubilities may be expressed by two independent curves and
`that the point where two curves meet is the pHmax (reproduced from Ref. [15]
`with permission).
`
`curves, one where the free base is the saturation or equilibrium
`species and the other where the salt is the equilibrium species.
`Essentially, the following equilibrium exists when a basic com-
`pound or its salt is dissolved in water:
`
`þ þ H2O fKa B þ H3O
`

`
`BH
`
`or
`
`
`½Ka ¼ B½ Š H3O
`




`BH
`

`
`ð1Þ
`
`ð2Þ
`
`Š ¼ B½ Š
`
`þ BH½
`
`Þ ¼ B½ Š
`

`
`

`¼ B½
`s 1 þ 10pKapH
`where the subscript “s” represents the saturation species. On the
`other hand, when the salt
`is the saturation species,
`the
`equilibrium solubility at a particular pH may be expressed by:
`

`ST; salt pH bpH max
`
`Þ ¼ BH½
`

`¼ BH

`

`
`s
`
`
`½þ B½ Š ¼ BH
`
`

`Šs 1 þ Ka
`

`
`

`H3O
`Šs 1 þ 10pHpKa
`
`ð4Þ
`
`
`
`The two independent curves mentioned above may be
`obtained by varying hydrogen ion concentrations (or pH)
`in Eqs. (3) and (4), and the point where the curves intersect
`the pH of maximum solubility. This is
`is called pHmax,
`shown schematically in Fig. 1, where the solubility profile at
`a pH higher than the pHmax is represented by Eq. (3), while
`Eq. (4) represents the solubility profile below pHmax. If
`the solid phase that is in equilibrium with a solution is ana-
`lyzed, it would be the free base at pH N pHmax and the salt
`at pH b pHmax. Only at pHmax, which theoretically represents
`
`Fig. 3. Schematic representation of the pH–solubility profile of an acidic drug
`indicating that the solubility may be expressed by two independent curves and
`that the point where the two curves meet is the pHmax (reproduced from Ref. [15]
`with permission).
`
`Merck Exhibit 2193, Page 3
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`606
`
`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`organic counterion. The relevant equations below and above
`pHmax are given below [25]:
`
`
`s 1 þ Ka

`H3O
`

`ST; acid pHbpH max
`
`Þ ¼ AH½
`¼ AH½
`
`s
`


`Š ¼ AH½
`þ A½
`
`
`

`s 1 þ 10pHpKa
`

`ST; salt pH N pH max
`
`Š ¼ A½
`
`þ AH½

`Þ ¼ A½
`
`
`

`
`Šs 1 þ 10pKapH
`¼ A
`
`s
`
`
`


`ð5Þ
`
`
`


`
`Šs 1 þ H3O
`
`Ka
`ð6Þ
`
`As indicated in Fig. 3, the solid phase in equilibrium with a
`saturated solution at pH b pHmax is the free acid and the solid
`phase at pH N pHmax is the salt; only at pHmax, both forms coexist.
`Interconversion from the salt to the free acid form or vice versa
`may occur if the pH shifts from one side of the pHmax to the
`other. There are numerous reports in the literature indicating that
`Eqs. (5) and (6) are, in general, followed for solubilities of free
`acids and their salts, respectively [22,23,26–29]. In all cases, salts
`had higher solubilities than their corresponding free acids,
`although solubilities of different salt forms of a particular acid
`could vary.
`
`2.3. Effect of counterion on salt solubility
`
`Salt-forming agents used to prepare salts, such as acids to
`form salts of basic drugs and bases to form salts of acidic drugs,
`exert influences on salt solubility by exerting common-ion
`effects in solution. This may be seen in Fig. 2, where solubilities
`of methanesulfonate, hydrochloride and phosphate salts of
`haloperidol decreased gradually at pH below 2.5. This is due to
`the common-ion effect since the acids used to lower pH
`generated excess counterions.
`The common-ion effect may be explained by the following
`equilibrium that exists below pHmax for the salt of a basic drug:




`
`þ X½

`ð7Þ
`f BH
`

`
`BH
`
`
`
`X
`
`solid
`

`
`s
`
`where (BH+X−)solid denotes undissolved solid salt that is in
`equilibrium with solution, [BH+]s is the salt solubility, and [X−]
`is the counterion concentration. The apparent solubility product
`(Ksp′ ) can be derived from Eq. (7) as follows:
`
`K Vsp ¼ BH½



`s X
`p
`ffiffiffiffiffiffiffi
`In the absence of excess counterion, [BH+]s = [X−], and there-
`K Vsp
`. Under such a condition, the solubility of a
`fore, solubility =
`salt remains unchanged as seen in the flat region of salt solubility in
`Fig. 2. On the other hand, if a significant amount of excess
`counterion is used either to lower pH or as a formulation adjuvant
`in dosage form (e.g., in adjusting ionic strength, tonicity, etc.), a
`major decrease in solubility may be observed, according to:


`
`¼ Ksp= X½


`BH
`
`ð8Þ
`
`ð9Þ
`

`
`s
`
`Streng et al. [30] studied the combined effect of the addition
`of NaCl and HCl on aqueous solubility of the HCl salt of a basic
`drug; the solubility in the relatively flat region of the pH–
`solubility profile (pH 3 to 6) decreased by a factor of 3 when
`0.05 M NaCl was added to the solution, while at pH below 3 the
`solubility further decreased due to the effect of Cl− ion as-
`sociated with HCl added to adjust pH. Similarly, in developing a
`liquid formulation for the sodium salt of an acidic drug,
`Serajuddin et al. [31] observed a decrease in solubility from
`7.8 mg/mL to 1.1 mg/mL with the addition of 0.1 M NaCl to
`adjust the ionic strength of solution. The common-ion effect
`also has a major influence on solubility and dissolution rates of
`salts in the GI tract, where the solubility of HCl salts are
`particularly sensitive to the presence of chloride ion [24].
`The overall impact of counterions on salt solubility depends
`on the magnitude of Ksp value. According to Eq. (9), for an
`equal change in [X−],
`the common-ion effect will be less
`pronounced in a salt of higher Ksp (i.e. higher solubility) than in
`a salt with lower Ksp (i.e. lower solubility). For example, the
`aqueous solubility of
`tiaramide HCl at 37 °C remained
`practically constant around 200 mg/mL (∼0.5 M) during the
`lowering of pH from 4.0 to 1.6 by the addition of HCl, since, as
`compared to the drug concentration, changes in the chloride ion
`concentration during the pH adjustment were negligible.
`Further, the solubility of tiaramide HCl decreased by just 25%
`to ∼150 mg/mL at pH 1. In contrast, there are numerous reports
`in the literature indicating drastic common-ion effects on salts
`having relatively low aqueous solubilities [18,21]; three such
`examples demonstrating major impacts of maleate and chloride
`ions on solubilities of a maleate salt [32] and two hydrochloride
`salts [33,34], respectively, at low pH are shown in Fig. 4. Since,
`as mentioned earlier, most compounds currently synthesized in
`drug discovery laboratories have poor aqueous solubilities and,
`as a consequence, their salt forms are also found to have
`relatively low aqueous solubilities, an investigation of potential
`impacts of common ions is critically important in salt selection
`and during dosage form development.
`
`2.4. Effects of solubility, pKa and Ksp on pHmax
`
`The concept of pHmax is an important one in the physical
`chemistry of salts. It is apparent from Figs. 1 and 3 that pHmax
`plays a major role in determining whether a salt would be
`formed or not, and, in case it is formed, whether it would remain
`‘as is’ or would convert to the corresponding free acid or base
`form. As mentioned previously, it is only at the pHmax that both
`forms could coexist. Therefore, at the pHmax, both Eqs. (3) and
`(4) can be valid for the solubility of a basic drug and, similarly,
`both Eqs. (5) and (6) can be valid for the solubility of an acidic
`drug. Bogardus and Blackwood [18] proposed that, for a basic
`drug, the saturation solubilities of free base and its salt form
`may be set equal at pHmax, and solving the relevant equations
`for pHmax, they derived the following relationship:
`
`pH max ¼ pKa þ log
`
`sffiffiffiffiffiffiffi
`p
`B½ Š
`
`Ksp
`
`ð10Þ
`
`Merck Exhibit 2193, Page 4
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`607
`
`It is evident from Fig. 5 that a stronger basicity (higher pKa), a
`higher intrinsic solubility and a lower salt solubility will favor salt
`formation for a basic drug by increasing pHmax. Analogous
`relationships may also be derived for the salt formation of an
`acidic drug where an increase in S0 and decreases in pKa and salt
`solubility will decrease pHmax and, therefore, favor salt formation.
`
`2.5. Deviation of pH–solubility interrelationship from ideality
`
`Organic compounds often undergo self-association in solu-
`tion because of their amphiphilic nature [35,36]. Indeed, bile
`salts are great examples of how organic compounds exhibit
`surface activity and undergo self-association in aqueous solu-
`tions because of their amphiphilic properties [37]. It has been
`reported that salt forms of many drug molecules undergo similar
`aggregation in solution [38–41]. Because of self-aggregation,
`activities of saturated solutions of many salts and even non-salts
`are lower than their measured concentrations in solution, re-
`sulting in non-ideal pH–solubility behavior. An example of
`
`Fig. 4. Typical pH–solubility profiles of poorly water-soluble basic drugs:
`(a) the solubility profile of a compound with intrinsic solubility (S0) of 2 μg/mL
`and a pKa of 6.3, for which a pHmax of ∼3.4 and a common-ion effect below
`pHmax were observed when the pH was lowered using maleic acid (reproduced
`with permission from Ref. [32]); (b) solubility profile of a compound with S0 of
`3.4 μg/mL and pKa of 5.7, for which pHmax of 3.2 and common-ion effect below
`pHmax were observed when the pH was lowered using HCl; and (c) the solubility
`profile of a base having S0 of b0.0001 μg/mL (below detection limit) and
`estimated pKa in the range of 5.5 to 6.0, for which the pH was adjusted by HCl
`and the hydrochloride salt did not have acceptable properties for further
`development due to low pHmax (∼1.5), low salt solubility (0.1 mg/mL at pHmax)
`and strong common-ion effect (reproduced with permission from Ref. [34]).
`
`Pudipeddi et al. [16] depicted the influence of S0 (or [B]s),
`pKa and Ksp on pHmax, according to Eq. (10), by using Fig. 5,
`where:
`
`a) an increase in pKa by one unit increases the pHmax by one
`unit;
`b) an increase in intrinsic solubility, S0, of the base by one order
`of magnitude increases pHmax by one unit; and
`c) a decrease in salt solubility (Ksp) by one order of magnitude
`increases pHmax by one unit.
`
`Fig. 5. Effects of (a) pKa, (b) S0 and (c) Ksp on pHmax (reproduced from Ref. [16]
`with permission).
`
`Merck Exhibit 2193, Page 5
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`608
`
`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`increase in activity coefficients of salts in solution [18,21,22]. In
`other words, solubilities of HCl salts decreased not only due
`to common-ion effect, but also due to salting out effects that
`decreased self-association [42].
`Bergstrom et al. [43] reported that experimentally determined
`solubility profiles of 25 amine drugs differed substantially from
`theoretical pH–solubility profiles generated by using the
`Henderson–Hasselbach (HH) equation. The HH equation used
`is essentially the same as Eq. (3) described earlier for the pH–
`solubility profile of a base. This equation would not describe the
`solubility of a salt form and, therefore, a deviation in solubility
`profile at pH b pHmax was expected. Some of the deviations
`observed at pH N pHmax could be due to self-association of
`dissolved species, especially at or near pHmax. The accuracy of a
`theoretical pH–solubility profile also depends on accuracies of
`intrinsic solubility and pKa values used in Eq. (3). The work of
`Bergstrom et al. [43] highlights the importance of accurate S0
`and pKa values; otherwise, the theoretical profiles generated may
`fail to appropriately predict the experimental behavior.
`
`2.6. Structure–solubility relationships
`
`As shown earlier in Fig. 2, aqueous solubilities of halo-
`peridol salts differed depending on salt-forming agents used.
`There are numerous other reports in the literature presenting
`such results for both acidic and basic drugs [25,30,44–48]. For
`example, Streng et al. [30] reported that solubilities of ter-
`fenadine salts formed with phosphoric acid, hydrochloric acid,
`methanesulfonic acid and lactic acid showed up to 10-fold
`differences, ranging from 0.5 mg/mL to 5 mg/mL. It may be
`noted that acidities and structures of acids used to form the salts
`in this study differed greatly. On the other hand, Serajuddin [33]
`reported for a basic drug, avitriptan, that solubilities of salts
`could be similar if structurally similar acids are used (Table 1).
`Although avitriptan is a dibasic compound with pKa values of
`8.0 and 3.6 [15], it formed mono-salts with all acids tested,
`except for HCl, which was also able to form a dihydrochloride
`salt. The solubility differed significantly only for the hydro-
`chloride salts. The results obtained by O'Connor and Corrigan
`[29] for diclofenac salts with a series of structurally similar
`amines, however, differed from the observation of Serajuddin
`[33] that similar counterions might provide similar solubilities;
`their values differed by a factor as much as N100.
`Anderson and Flora [13]
`indicated that no predictive
`structure–solubility relationships for pharmaceutical salts have
`yet been established. They, however, suggested that
`for
`
`Table 1
`Aqueous solubility of mono-salts of avitriptan containing various counterions
`
`Acid used
`
`pKa of acid
`Solubility (mg/mL at 25 °C)
`−6.1
`3.4 (9.0) a
`HCl
`−1.2
`Methanesulfonic acid
`16.3
`Tartaric acid
`3.0, 4.4
`14.7
`Lactic acid
`3.9
`15.2
`Succinic acid
`4.2, 5.6
`16.1
`Acetic acid
`4.8
`16.5
`a Value in parentheses is for the dihydrochloride salt.
`
`Fig. 6. pH–solubility profile of papaverine hyrochloride determined by using HCl
`or NaOH, as necessary, indicating supersaturation around pHmax and common-ion
`effect at low pH. The broken line shows the theoretical solubility profile determined
`based on S0 and pKa, and the positive deviation of experimental profile indicates
`self-association (reproduced from Ref. [19] with permission).
`
`such non-ideality is shown in Fig. 6, in which the saturation
`solubility of papaverine hydrochloride at or below pHmax
`exhibits higher values than the theoretical profile. There are also
`numerous examples where metastable, supersaturated solutions
`are formed at or near pHmax [13,19–22]. Supersaturated solu-
`tions are formed more often when free base or free acid is used
`as the starting material in the phase solubility study due to
`‘kinetic barriers’ in the transformation of free species to the salt
`form. Such kinetic barriers may also be present in the con-
`version of salts to free species, but they are relatively less
`pronounced. Serajuddin and Mufson [20] reported that
`the
`solubility of papaverine free base at 37 °C increased gradually
`when the pH was decreased by adding HCl to a suspension until
`the pH reached 4. Then, at an almost constant pH of 4 ± 0.1, the
`solubility increased from b10 mg/mL to N120 mg/mL; the
`solubility kept on increasing with the addition of HCl as long as
`solid base was present to dissolve into solution. Although it was
`known that the aqueous solubility of papaverine hydrochloride
`was only 40 mg/mL, there was no sign of precipitation of the
`supersaturated solution to the salt from even at a concentration
`above 120 mg/mL, and it was only after nucleation of the
`supersaturated solution by adding a few crystals of papaverine
`HCl that the precipitation of the salt ensued and the solubility
`dropped to the level of the salt form. With a similar addition of
`HCl to a phenazopyridine aqueous suspension, the solubility of
`the free base at pHmax was observed to reach at least three times
`higher than that of phenazopyridine HCl before precipitation of
`the salt form ensued [21,22]. Such a supersaturation and the
`deviation from the ideal pH–solubility relationship were
`attributed to self-association of drug in solution.
`The aggregation of drug in solution does not occur only at the
`pHmax. Ledwidge and Corrigan [22] demonstrated that salts of
`both basic and acidic drugs may self-associate and exhibit
`surface activity at a wide range of pH. For hydrochloride salts,
`the addition of an excess of chloride ion during the pH ad-
`justment showed a decrease in apparent Ksp values, which was
`attributed to a decrease in self-association and the consequent
`
`Merck Exhibit 2193, Page 6
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`A.T.M. Serajuddin / Advanced Drug Delivery Reviews 59 (2007) 603–616
`
`609
`
`understanding the effect of a series of salt-forming counterions
`on aqueous solubility, contributions of the counterions to crystal
`lattice energies and solvation energies should be considered. In
`dissolving a salt in water, the molar free energy of solution,
`▵Gsoln, may be represented by
`ð11Þ
`DGsoln ¼ DGcation þ DGanion DGlattice
`where ▵Gcation and ▵Ganion are molar free energies of hydration
`of the salt cationic and anionic species, respectively, and ▵Glattice
`is the crystal lattice free energy. Thus, the overall effect of
`counterions on salt solubility will depend on whether hydration
`energies or lattice energies are most sensitive to changes in salt
`structure. By analyzing data for alkali and alkaline earth metal
`salts of several carboxylic acids, the authors suggested that a
`quantitative trend exists where the solubility increases with an
`increase in anion or cation charge and decreases with an increase
`in ionic radius. A general trend was also observed where solubility
`of various ammonium salts of an acidic drug, flurbiprofen,
`increased with a decrease in melting point, indicating that crystal
`lattice energy plays an important role in salt solubility.
`Several other investigations also attempted to establish
`relationships between salt forms and their aqueous solubilities
`[28,29,44–46]. However, no general predictive relationships
`could be obtained. For example, it was reported that solubilities
`of diclofenac salts with several structurally related primary
`amines varied by a factor of as much as 100, and they did not
`show dependence on any one parameter, but on a combination
`of factors like salt crystal
`lattice and pH of saturated salt
`solution [29].
`
`2.7. Effect of organic solvent on salt solubility
`
`The pH–solubility principles described in this section are
`applicable to aqueous solutions. However, as reported by
`Wermuth and Stahl [49], pharmaceutical salts are usually
`synthesized either from organic solvents or from organic–water
`cosolvents. No systematic studies on the solubility of acidic and
`basic drugs in such solvents as a function of added counterions to
`form salts have been reported in the literature.
`Organic solvents are used not only in the preparation of salts;
`they are also used in parenteral and other liquid dosage forms.
`Organic solvents may influence the solubility of a drug candidate
`by (a) increasing solubility of unionized species (S0), (b) de-
`creasing its protonation or ionization, and (c) decreasing solubility
`of the salt form [15]. Thus, as reported by Kramer and Flynn [17],
`an increase in S0 in an organic cosolvent will increase pHmax for a
`basic drug, thus favoring salt formation. This is, of course,
`assuming that the pKa value of the compound would remain
`unchanged. However, the presence of organic solvents may
`adversely affect drug ionization by suppressing the dielectric
`constant [50]. For example, Albert and Serjeant [51] reported that
`the pKa of an acid increased by ∼1 and that of a base decreased by
`∼0.5 in a 60:40 methanol–water mixture. As a consequence, any
`positive influence of organic solvents on salt formation may partly
`or fully be negated by the decreased ionization. The decreased salt
`solubility in the presence of organic solvents may influence drug
`
`produc

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