throbber
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / e j p s
`
`Commentary
`Generic substitution: The use of medicinal
`products containing different salts and
`implications for safety and efficacy
`
`R.K. Verbeeck 1, I. Kanfer
`Faculty of Pharmacy, Rhodes University, Artillery Road, Grahamstown 6140, South Africa
`
`, R.B. Walker
`
`∗
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 25 May 2005
`Received in revised form 31 October
`2005
`Accepted 4 December 2005
`Published on line 18 January 2006
`
`Keywords:
`Generic substitution
`Bioequivalence
`Pharmaceutical equivalence
`Therapeutic equivalence
`Pharmaceutical alternative
`
`In their quest to gain early entry of new generic products into the market prior to patent expi-
`ration, one of the strategies pursued by generic drug product manufacturers is to incorporate
`different salts of an approved active pharmaceutical ingredient (API) in a brand company’s
`marketed dosage form and subject such dosage forms to bioequivalence assessment. These
`initiatives present challenges to regulatory authorities where the decision to approve bioe-
`quivalent products containing such pharmaceutical alternatives must be considered in the
`light of safety and efficacy, and more particularly, with respect to their substitutability.
`This article describes the various issues and contentions associated with the concept of
`pharmaceutical alternatives, specifically with respect to the uses of different salts and the
`implications for safety, efficacy and generic substitution.
`© 2005 Elsevier B.V. All rights reserved.
`
`1.
`
`Introduction
`
`Most drugs are either weak organic acids or weak organic bases
`and can therefore exist as different salt forms. Although the
`active pharmaceutical ingredient (API) in these different salts
`is the same, each of these salts may be considered as being
`distinct chemical entities with their own chemical and bio-
`logical profiles which may lead to differences in their clinical
`efficacy and safety (Berge et al., 1977; Gould, 1986; Davies,
`2001; Stahl and Wermuth, 2002a). The term pharmaceutical
`alternatives is used in relation to different salts (or esters)
`of the same active substance in the EU Note for Guidance
`
`as well as in the FDA Guidance for Industry on Bioavailabil-
`ity and Bioequivalence Studies for Orally Administered Drug
`Products (EMEA, 2001; FDA, 2000). According to the EU guide-
`lines “medicinal products are pharmaceutical alternatives if
`they contain the same active moiety but differ in chemical
`form (salt, ester, etc.) of that moiety or in the dosage form or
`strength”. Similarly, the definition of pharmaceutical alterna-
`tives as stated in the FDA’s “Approved Drug Products with Ther-
`apeutic Equivalence Evaluations”, 24th edition (Orange Book,
`2004) is as follows: “Drug products are considered pharmaceu-
`tical alternatives if they contain the same therapeutic moiety,
`but are different salts, esters, or complexes of that moiety,
`
`∗
`
`Corresponding author. Tel.: +27 46 603 8382; fax: +27 46 636 1205.
`E-mail address: I.Kanfer@ru.ac.za (I. Kanfer).
`1 Present address: School of Pharmacy, UCL/PMNT 7369, Av. E. Mounier 73, Brussels, Belgium.
`0928-0987/$ – see front matter © 2005 Elsevier B.V. All rights reserved.
`doi:10.1016/j.ejps.2005.12.001
`
`Merck Exhibit 2196, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`2
`
`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`or are different dosage forms or strengths . . .”. In contrast
`to the issue of pharmaceutical alternatives, the Orange Book
`(2004) also defines the term, pharmaceutical equivalents, as
`follows: “Drug products are considered pharmaceutical equiv-
`alents if they contain the same active ingredient(s), are of the
`same dosage form, route of administration and are identi-
`cal in strength or concentration. Pharmaceutically equivalent
`drug products are formulated to contain the same amount
`of active ingredient in the same dosage form and to meet
`the same or compendial or other applicable standards (i.e.
`strength, quality, purity and identity), but they may differ in
`characteristics, such as shape, scoring configuration, release
`mechanisms, packaging, excipients (including colors, flavours
`and preservatives), expiration time and within certain limits,
`labelling”.
`According to both the FDA (2000) and EMEA (2001)
`guidelines, bioequivalence can be established between two
`medicinal products, which are pharmaceutical alternatives.
`However, the definition of therapeutic equivalence as given
`in the Orange Book (2004) precludes the substitutability of
`pharmaceutical alternatives, as follows: “Drug products are
`considered to be therapeutic equivalents only if they are phar-
`maceutical equivalents and if they can be expected to have
`the same clinical effect and safety profile when administered
`to patients under the conditions specified in the labelling”.
`On the other hand the European Agency for the Evaluation
`of Medicinal Products (EMEA) makes provision for medici-
`nal products which are either pharmaceutically equivalent
`or pharmaceutical alternatives to be declared as therapeu-
`tic equivalents, as follows: “In practice, demonstration of
`bioequivalence is generally the most appropriate method
`of substantiating therapeutic equivalence between medici-
`nal products which are pharmaceutically equivalent or phar-
`maceutical alternatives, provided they contain excipients
`generally recognised as not having an influence on safety
`and efficacy and comply with labelling requirements with
`respect to excipients” (EMEA, 2001). The immediately pre-
`ceding paragraph in the same EMEA guideline confound-
`ingly states that: “A medicinal product is therapeutically
`equivalent with another product if it contains the same
`active substance or therapeutic moiety and, clinically, shows
`the same efficacy and safety as that product, whose effi-
`cacy and safety has been established”. The issue is compli-
`cated by incorporation of the phrase “. . ., clinically, shows
`the same efficacy and safety as that product, whose effi-
`cacy and safety has been established”, in the definition.
`In our view this implies that therapeutic equivalence can-
`not be established between pharmaceutical alternatives on
`bioequivalence data alone. Hence, whereas pharmaceutically
`equivalent products can clearly be considered therapeuti-
`cally equivalent based on a bioequivalence study, additional
`preclinical and/or clinical data may be required for a pharma-
`ceutical alternative to be considered therapeutically equiva-
`lent.
`In this commentary, scientific facts/data will be pre-
`sented to show that establishing bioequivalence between
`oral drug products containing different salts of the same
`active substance, will usually not suffice to claim therapeutic
`equivalence and consequently substitutability/interchange-
`ability.
`
`Active pharmaceutical ingredients and
`2.
`their salts
`
`Converting an API to a particular salt form is a means of mod-
`ifying and sometimes optimising its physicochemical prop-
`erties (Stahl and Wermuth, 2002a,b). However, changing the
`salt form may also affect the biological properties of the
`drug and have significant implications for safety and tox-
`icity (Davies, 2001). The most appropriate salt form of an
`active moiety should ideally be selected at an early stage
`of the development of a New Chemical Entity (NCE) to opti-
`mise the characteristics of the final formulation. Indeed, dif-
`ferent salt forms of a particular API can differ markedly in
`physicochemical properties, such as solubility, hygroscopicity,
`stability, flowability, etc. In addition, the presence of impu-
`rities associated either with the route of synthesis of that
`particular salt or resulting as a consequence of instability and
`the formation of degradation products, can impart toxicity
`and/or undesirable biological activity quite different from the
`drug’s intended clinical use (Bastin et al., 2000; Byrn et al.,
`1995). Hence, it may therefore be possible that substitution
`of one salt form of an API for another can alter therapeu-
`tic efficacy, safety and/or quality. Unfortunately, there is no
`reliable way of predicting the influence of a particular salt
`species on the behaviour of the parent compound in dosage
`forms.
`It is estimated that half of all the active substances used
`in medicinal therapy are administered as salts, and salifica-
`tion of a drug substance has become an essential step in drug
`development (Balbach and Korn, 2004; Gardner et al., 2004).
`Selecting an appropriate salt form of an API is not only an
`important consideration in the early stages of new drug devel-
`opment (Bowker, 2002), it may also play a role in the develop-
`ment of generic drug products as illustrated by the example
`of amlodipine. This calcium channel blocker is marketed by
`Pfizer as the besylate salt (Norvasc®). Pfizer’s original patent
`on amlodipine besylate expired in 2003 but was extended until
`2007 to compensate for a lengthy review process by the FDA
`(Anon., 2004). Pfizer’s original patent attempted to protect both
`the chemical structure of amlodipine besylate and a series
`of other salts of amlodipine. Dr. Reddy’s Laboratories Limited
`developed a generic version of amlodipine in the form of the
`maleate salt and showed that their product (AmVazTM, Reddy
`Pharmaceuticals Inc.) was bioequivalent to Pfizer’s Norvasc®
`(Suh et al., 2004). Dr. Reddy’s Laboratories tried to obtain mar-
`keting authorization arguing that Pfizer’s patent extension
`did not apply to their version of the drug, i.e. amlodipine
`maleate. However, on February 27, 2004 The United States
`Court of Appeals for the Federal Circuit reversed the ear-
`lier New Jersey District Court’s dismissal of Pfizer’s patent
`infringement action against Dr. Reddy’s Laboratories’ generic
`version of Norvasc®, thus effectively preventing the generic
`version from entering the market (Anon.: Pfizer Inc. ver-
`sus Dr. Reddy’s Laboratories, www.ll.georgetown.edu/federal/
`judicial/fed/opinions/03opinions/03-1227.html, visited 05/23/
`05). A short discussion of the properties of amlodipine
`maleate, with particular emphasis on stability and subsequent
`effects on efficacy and safety is presented in Section 3.4 (vide
`infra).
`
`Merck Exhibit 2196, Page 2
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`3
`
`Apart from the legal issues, the important question to
`be answered is: what experiments and tests are required
`to ensure that a drug product containing a specific salt
`form of an API has comparable pharmacokinetic, pharma-
`cological, toxicological and safety profiles as the registered
`product containing an alternative salt form of the same
`active substance? Furthermore, what is the likelihood that
`pharmaceutical alternatives which have been shown to be
`bioequivalent will have different clinical safety and efficacy
`profiles?
`
`Development of generic drug products
`3.
`using an alternative salt of the same active
`moiety
`
`The following issues are important when considering whether
`alternative salt forms of the same active moiety can be
`considered therapeutically equivalent and hence have to be
`addressed when developing a generic drug product using an
`alternative salt form of the active substance.
`
`3.1.
`
`Solubility, dissolution and bioavailability
`
`Many examples can be found in the scientific literature
`showing that the water solubilities of alternative salt forms
`of the same active moiety can be quite different. The antide-
`pressant, trazodone, for example,
`is currently marketed
`as the hydrochloride salt. Ware and Lu (2004) prepared a
`number of alternative salts in an attempt to find a salt form
`of trazodone with lower aqueous solubility compared to
`trazodone hydrochloride. Among the salts selected for final
`evaluation, the tosylate and pamoate salts of trazodone
`were less water-soluble than the sulphate and hydrochloride
`salts. The tosylate salt showed the most interesting solu-
`bility profile with values ranging from 3 mg/ml at pH 1.0 to
`0.2 mg/ml at pH 12.0. This characteristic makes it the best
`candidate, compared to the other salts, for the development
`of a prolonged release oral trazodone product to improve
`patient compliance in the elderly. Because of the significantly
`lower (8–10-fold in the pH range 1–5) solubility of the tosylate
`salt compared to the marketed hydrochloride salt, the in vivo
`absorption rate of trazodone following oral administration of
`the tosylate salt may be significantly lower. Consequently, the
`two salts will probably be neither bioequivalent, i.e. having
`a similar rate and extent of absorption, nor therapeutically
`equivalent.
`Following oral administration as a solid dosage form, the
`dissolution rate of the active substance in the gastrointesti-
`nal juices is affected by its aqueous solubility. Therefore,
`solid dosage forms containing alternative salts of the same
`active substance may show different in vivo dissolution
`characteristics. According to the principles underlying the
`Biopharmaceutics Classification System,
`for active drug
`substances with a high intestinal permeability, the in vivo
`dissolution rate will determine the rate and in some cases
`also the extent of absorption (Amidon et al., 1995). For active
`substances with a low intestinal permeability and a relatively
`good aqueous solubility, however, in vivo dissolution is no
`longer the rate-limiting step in the absorption process and
`
`differences in aqueous solubility and dissolution are usu-
`ally not important determinants of bioavailability. Human
`bioequivalence studies comparing salt forms of basic drugs
`have been rather limited and none of them have reported
`significant differences in bioavailability between different salt
`forms due to differences in their aqueous solubilities (Engel et
`al., 2000). Lin et al. (1972), for example, reported no enhance-
`ment in bioavailability when salts of a basic antihypertensive
`agent,
`1-(2,3-dihydro-5-methoxybenzo[b]furan-2-ylmethyl)-
`4-(o-methoxyphenyl)piperazine, having significantly different
`intrinsic dissolution rates, were compared. Walmsley et al.
`(1986) also indicated that they did not observe a difference
`in the extent of bioavailability between oxalate and citrate
`salts of naftidrofuryl, while Jamuludin et al. (1988) saw no
`significant differences in Cmax, Tmax, or AUC of quinine
`following oral administration of the hydrochloride, sulphate
`and ethyl carbonate salts of this antimalarial to healthy
`volunteers. Consequently, it may be concluded that an in
`vivo bioequivalence study is absolutely necessary if thera-
`peutic equivalence between alternative salts of the same
`active drug molecule is being claimed, except when both
`salts are highly soluble and highly permeable, i.e. BCS class
`I compounds. In that case a BCS-based waiver for an in
`vivo BE study for an immediate release oral dosage form
`which exhibits rapid in vitro dissolution can be requested,
`provided a number of additional conditions are met (FDA,
`2000).
`
`3.2.
`
`Toxicity
`
`Toxicity associated with the salt of an active drug molecule
`may be due to the conjugate anion or cation used to form
`the salt (Berge et al., 1977; Stahl and Wermuth, 2002b). For
`example, the nephrotoxicity of pravadoline maleate, which
`was reported to cause renal tubular lesions in the dog, has
`been shown to be the result of the formation of maleic acid
`from the maleate anion (Everett et al., 1993). The need to evalu-
`ate the safety profile of the salt-forming agent depends largely
`on its chemical nature, its biological characteristics, whether
`the agent has been used in other medicinal products, foods
`and beverages or not, as well as the relative ratio of the salt-
`forming component to the active substance. Toxicity studies
`are required for a new salt form of an active substance when
`the salt of that active substance has been prepared by using a
`new salt-forming agent with little or no information on its tox-
`icity profile. Toxicity studies on the salt-forming agent alone
`are also necessary. Monographs on 68 salt-forming acids and
`27 salt-forming bases have been published in the Handbook
`of Pharmaceutical Salts: Properties, Selection and Use, edited by
`Stahl and Wermuth (2002a) as well as a comprehensive list
`of salt-forming acids and bases with information regarding
`their safety/toxicity (Stahl and Wermuth, 2002b; Wermuth,
`2002).
`Potentially toxic chemical impurities formed during the
`preparation of a specific salt of an API may also explain dif-
`ferences found in the toxicity profiles of various salt forms
`of an active drug molecule. It is therefore necessary to eval-
`uate the toxic potential of all impurities found during the
`synthesis of a specific salt form (Bauer et al., 1998). For
`example, methane sulfonic acid is used in the formation of
`
`Merck Exhibit 2196, Page 3
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`4
`
`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`methane sulfonates (also called mesylate salts) of active basic
`drug molecules, such as pergolide, nelfinavir, imatinib and
`amlodipine. Benzene and toluene sulfonates (besylates and
`tosylates, respectively), have also been prepared. Recently, the
`potential health hazards of trace amounts of mesylate esters,
`including methyl methanesulfonate, ethyl methanesulfonate
`and isopropyl methanesulfonate, in pharmaceuticals have
`attracted the attention of health authorities (Anon., 2000).
`These impurities could arise from the reaction of methane
`sulfonic acid with solvents, such as methanol, ethanol and iso-
`propyl alcohol during the manufacture of the mesylate salts
`of active substances. These mesylate esters are known to be
`potent mutagenic, carcinogenic and teratogenic compounds
`(Sega et al., 1986; Morris et al., 1994). In general, it can be
`concluded that when the routes to synthesize or prepare alter-
`native salt forms of the same active moiety result in different
`chemical by-products, the toxic potential of these impurities
`should be evaluated by preclinical testing for each salt form
`synthesized/prepared.
`The specific salt form of an active substance may also
`affect tolerability. Gastrointestinal irritation and ulceration,
`for example, may be dependent upon the aqueous solubil-
`ity and dissolution rate of different salt forms administered
`by the oral route. Olovson et al. (1986) tested the ulcero-
`genic effect of five different salts of alprenolol against placebo
`in a porcine oesophageal test model. The salts with high
`water solubility, such as the hydrochloride and the fumarate,
`gave rise to the highest plasma concentrations of alprenolol
`and evoked serious oesophageal lesions, while the salts with
`low solubility – the benzoate, maleate and sebacate – had
`no irritant effect on the oesophagus. The plasma levels of
`alprenolol were much higher following administration of
`alprenolol hydrochloride in the oesophagus than after an iden-
`tical intraduodenal dose of the same salt possibly because of
`the avoidance of first-pass metabolism during oesophageal
`absorption.
`
`3.3.
`
`Polymorphism
`
`The solid-state properties of a molecule, as well as its proper-
`ties in solution, can be modified by salt formation. Selecting a
`salt suitable for a certain route of administration or a particu-
`lar dosage form of a drug substance requires that all the rele-
`vant solid-state properties of the salt candidates be thoroughly
`investigated. Polymorphism is frequently a critical point in
`determining preferences for one salt or another (Balbach and
`Korn, 2004; Bowker, 2002). Polymorphism can be defined as the
`ability of a drug substance to exist as two or more crystalline
`phases that have different arrangements and/or conforma-
`tions of the molecules in the crystal lattice. Polymorphism is
`a widespread phenomenon observed in more than half of all
`active drug substances. The most critical issue related to drug
`substance polymorphism is equilibrium solubility which is
`an important determinant of dissolution rate and which may
`affect the bioavailability following oral administration of the
`active substance (Huang and Tong, 2004). Clearly, if polymor-
`phism has an effect on the bioavailability of a drug substance,
`a bioequivalence study between two formulations containing
`different polymorphs of the same drug should reveal those
`effects.
`
`Stability and formulation/production
`3.4.
`considerations
`
`As mentioned before, the different salt forms of an active
`drug moiety can vary in a number of physicochemical char-
`acteristics including hygroscopicity. Increased hygroscopic-
`ity may reduce stability of the active drug moiety, even in
`a pharmaceutical dosage form, such as tablets, especially
`when the active drug moiety is susceptible to hydrolytic
`degradation. In addition, thermal stability and degradation
`pathways may be different for alternative salt forms of the
`same active moiety possibly requiring the need to evaluate
`new degradation products by using appropriate toxicological
`studies.
`Amlodipine maleate provides an interesting example
`where instability of this particular salt results in the for-
`mation of a degradation product, which has significant
`implications for safety and toxicity. The maleate salt of
`amlodipine, unlike the besylate salt, suffers from intrinsic
`chemical instability which results in the formation of N-(2-{[4-
`(2-chlorophenyl)-3-(ethoxycarbonyl)-5-(methoxycarbonyl)-6-
`methyl-1,4-dihydro-2-pyridyl]methoxy}ethyl) aspartic acid,
`an impurity with demonstrated biological activity. It is formed
`by an intramolecular reaction of the unsaturated maleic acid
`with the primary amine group of amlodipine. This compound
`has been shown to possess a distinctly different biological
`profile to amlodipine itself (Amlodipine Citizen Petition,
`http://www.fda.gov/ohrms/dockets/dailys/03/Sept03/090303/
`03p-0408-cp00001-08-Tab-G-vol3.pdf, visited 05/23/05). Hence,
`the maleate salt of amlodipine cannot be considered to be
`therapeutically equivalent to the besylate salt since the latter
`does not have this additional clinical effect. The consequences
`of the presence of the biologically active impurity associated
`with amlodipine maleate therefore militates against generic
`substitution between maleate and besylate salts even if
`bioequivalence can be demonstrated. Whereas low levels of
`this impurity may not result in serious clinical consequences,
`the instability of the amlodipine maleate salt suggests that
`relatively high levels would likely result following the manu-
`facture of dosage forms and on prolonged storage. However,
`a case could be made to suggest interchangeability and thus
`permit generic substitution if a stabilised formulation of
`amlodipine maleate is used to show bioequivalence between
`the maleate and besylate salts. Such stabilized formulations
`have been described in a recent patent (Bilotte et al., 2002)
`where it is claimed that the formation of amlodipine aspartate
`can be prevented.
`The choice of a particular salt form can have a profound
`effect on the physicochemical properties, which are critical for
`the optimal formulation of the dosage form and large-scale
`manufacturing. The melting point of a particular salt often
`plays an important role. Generally, drugs with low melting
`points exhibit plastic deformation which can result in caking
`and aggregation of the API which can alter flow properties and
`compression characteristics and subsequently impact nega-
`tively on dose uniformity, friability, disintegration and disso-
`lution rate of solid dosage forms. The formation of plastic
`materials can create problems for size reduction and tablet
`processing due to melting and deposition of drug on milling
`equipment and film formation on tabletting punches with
`
`Merck Exhibit 2196, Page 4
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`5
`
`deleterious consequences for the bulk manufacture of tablets
`(Florence and Attwood, 1988).
`
`4.
`
`Regulatory requirements
`
`The health authorities of the European Union as well as those
`of the USA consider alternative salts of approved drug sub-
`stances as NCEs (Asche et al., 2002). However, the application
`to register medicinal products containing an alternative salt
`of an approved active substance as a generic product may
`be facilitated, under certain conditions, by the use of previ-
`ous knowledge on and clinical experience gained with the
`active moiety approved as a different salt form. Therefore, in
`many cases of salt changes or development of a generic drug
`product on the basis of an alternative salt form of the active
`moiety already marketed, an abbreviated or abridged applica-
`tion may be submitted as long as evidence can be provided
`that the alternative salt form does not lead to a change in the
`pharmacokinetics of the active moiety, nor in its pharmaco-
`dynamic and/or toxicity characteristics, which could change
`the safety/efficacy profile. Notwithstanding the above, in the
`USA, pharmaceutical alternatives which have been shown to
`be bioequivalent to an approved reference product containing
`a different salt and/or dosage form, would not be considered
`to be therapeutically equivalent and generic substitution of
`such products is therefore not permitted.
`
`5.
`
`Conclusions
`
`According to the CPMP Note for Guidance on the Investigation
`of Bioavailability and Bioequivalence, demonstration of bioe-
`quivalence is the most appropriate method of substantiating
`therapeutic equivalence between medicinal products which
`are pharmaceutically equivalent or pharmaceutical alterna-
`tives, such as different salt forms of the same active moiety
`(EMEA, 2001). If bioequivalence between two different salts of
`the same active moiety has been demonstrated, it is clear that
`any differences in physicochemical properties, such as solu-
`bility, between the two salts do not have any significant effect
`on the in vivo bioavailability of the active moiety. However,
`this does not suffice to conclude that these alternative salt
`forms are therapeutically equivalent. Therapeutic equivalence
`between two medicinal products not only implies the same
`efficacy but also the same safety profile. The issues raised
`above related to the possible difference in toxicity and sta-
`bility of two different salt forms of the same active moiety,
`demonstrate that an alternative salt form may have to undergo
`toxicological evaluation, in addition to a valid BE study show-
`ing in vivo bioequivalence, before therapeutic equivalence, for
`example, to a different (marketed) salt form of the same active
`moiety, can be accepted.
`
`r e f e r e n c e s
`
`Amidon, G.L., Lennern ¨as, H., Shah, V.P., Crison, J.R., 1995. A
`theoretical basis for a biopharmaceutic drug classification:
`the correlation of in vitro drug product dissolution and in
`vivo bioavailability. Pharm. Res. 12, 413–420.
`
`Anon., 2000. Alkyl mesylate (methanesulphonate) impurities in
`mesilate salts. Pharmeuropa 12, 27.
`Anon., 2004. Court reversal in favour of Pfizer. Nat. Rev. Drug
`Discov. 3, 290.
`Asche, H., Kondo, J.-I., Ajayi, F.O., 2002. Regulatory aspects of
`drug salts. In: Stahl, P.H., Wermuth, C.G. (Eds.), Handbook of
`Pharmaceutical Salts: Properties, Selection and Use.
`Wiley-VCH, Winheim, pp. 237–247.
`Balbach, S., Korn, C., 2004. Pharmaceutical evaluation of early
`development candidates “the 100 mg approach”. Int. J.
`Pharm. 275, 1–12.
`Bastin, R.J., Bowker, M.J., Slater, B.J., 2000. Salt selection and
`optimisation procedures for pharmaceutical new chemical
`entities. Org. Proc. Res. Dev. 4, 427–435.
`Bauer, M., de Leede, L., Van Der Waart, M., 1998. Purity as an
`issue in pharmaceutical research and development. Eur. J.
`Pharm. Sci. 6, 331–335.
`Berge, S.M., Bigly, L.D., Monkhouse, D.C., 1977. Pharmaceutical
`salts. J. Pharm. Sci. 66, 1–19.
`Bilotte, A., Pavitt, S., Pettman, A.J., Smith, Z.E.F., 2002. Stabilized
`Amlodipine Maleate Formulations. Pfizer Ltd., European
`Patent No. EP1266654.
`Bowker, M.J., 2002. A procedure for salt selection and
`optimisation. In: Stahl, P.H., Wermuth, C.G. (Eds.), Handbook
`of Pharmaceutical Salts: Properties, Selection and Use.
`Wiley-VCH, Winheim, pp. 161–189.
`Byrn, S., Pfeiffer, R., Ganey, M., Hoiberg, C., Poochikian, G., 1995.
`Pharmaceutical solids: a strategic approach to regulatory
`considerations. Pharm. Res. 12, 945–954.
`Davies, G., 2001. Changing the salt, changing the drug. Pharm.
`J. 266, 322–323.
`EMEA, 2001. Note for Guidance on the Investigation of
`Bioavailability and Bioequivalence, Committee for
`Proprietary Medicinal Products (CPMP). European Agency for
`the Evaluation of Medicinal Products, London, UK.
`Engel, G.L., Farid, N.A., Faul, M.M., Richardson, L.A., Winneroski,
`L.L., 2000. Salt form selection and characterization of
`LY333531 mesylate monohydrate. Int. J. Pharm. 198, 239–247.
`Everett, R.M., Descotes, G., Rollin, M., Greener, Y., Bradford, J.C.,
`Benziger, D.P., Ward, S.J., 1993. Nephrotoxicity of pravadoline
`maleate (WIN 48098-6) in dogs: evidence of maleic
`acid-induced acute tubular necrosis. Fundam. Appl. Toxicol.
`21, 59–65.
`FDA, 2000. Guidance for Industry: Bioavailability and
`Bioequivalence Studies for Orally Administered Drug
`Products—General Considerations. Center for Drug
`Evaluation Research, Food and Drug Administration, WA,
`USA.
`Florence, A.T., Attwood, D., 1988. Physicochemical Principles of
`Pharmacy, second ed. Macmillan Press, London.
`Gardner, C.R., Almarsson, O., Chen, H., Morissette, S., Peterson,
`M., Zhang, Z., Wang, S., Lemmo, A., Gonzales-Zugasti, J.,
`Monagle, J., Marchionna, J., Ellis, S., McNulty, C., Johnson, A.,
`Levinson, D., Cima, M., 2004. Application of high throughput
`technologies to drug substance and drug product
`development. Comp. Chem. Eng. 28, 943–953.
`Gould, P.L., 1986. Salt selection for basic drugs. Int. J. Pharm.
`33, 201–217.
`Huang, L.-F., Tong, W.-Q., 2004. Impact of solid state properties
`on developability assessment of drug candidates. Adv. Drug
`Deliv. Rev. 56, 321–334.
`Jamuludin, A., Mohamad, M., Navartnam, V., Selliah, K., Tan,
`S.C., Wernsdorfer, W.H., 1988. Relative bioavailability of the
`hydrochloride, sulphate and ethyl carbonate salts of
`quinine. Br. J. Clin. Pharmacol. 25, 261–263.
`Lin, S.L., Lachman, L., Schwartz, C.J., Huebner, C.F., 1972.
`Preformulation investigation. I: relation of salt forms and
`biological activity of an experimental antihypertensive. J.
`Pharm. Sci. 61, 1418–1422.
`
`Merck Exhibit 2196, Page 5
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`6
`
`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 2 8 ( 2 0 0 6 ) 1–6
`
`Morris, S.M., McGarrity, L.J., Domon, O.E., Chen, J.J., Hinson,
`W.G., Bucci, T.J., Warbritten, A.R., Casciano, D.A., 1994. The
`role of programmed cell death in the toxicity of the
`mutagens, ethyl methanesulfonate and
`(cid:2)
`N-ethyl-N
`-nitrosourea, in AHH-1 human lymphoblastoid
`cells. Mutat. Res. 306, 19–34.
`Olovson, S.-G., Havu, N., Reg ˚ardh, C.-G., Sandberg, A., 1986.
`Oesophageal ulcerations and plasma levels of different
`alprenolol salts: potential implications for the clinic. Acta
`Pharmacol. Toxicol. 58, 55–60.
`Orange Book, 2004. Approved Drug Products with Therapeutic
`Equivalence Evaluations, 24th ed. Food and Drug
`Administration, USA.
`Sega, G.A., Sluder, A.E., McCoy, L.S., Owens, J.G., Generoso, E.E.,
`1986. The use of alkaline elution procedures to measure
`DNA damage in spermiogenic stages of mice exposed to
`methyl methanesulfonate. Mutat. Res. 159, 55–63.
`Stahl, H., Wermuth, C.G. (Eds.), 2002a. Handbook of
`Pharmaceutical Salts: Properties, Selection and Use.
`Wiley-VCH, Weinheim.
`
`Stahl, P.H., Wermuth, C.G., 2002b. Monographs on acids and
`bases. In: Stahl, P.H., Wermuth, C.G. (Eds.), Handbookof
`Pharmaceutical Salts: Properties, Selection and Use.
`Wiley-VCH, Weinheim, pp. 265–327.
`Suh, S.S., Mohan, M.S., Davis, M.W., 2004. Amlodipine:
`pharmacokinetics of the maleate vs. besylate salts. Am. J.
`Hypertens. 17, 250.
`Walmsley, L.M., Taylor, T., Wilkinson, P.A., Brodie, R.R.,
`Chasseaud, L.F., Alun-Jones, V., Hunter, J.O., 1986. Plasma
`concentrations and relative bioavailability of naftidrofuryl
`from different salt forms. Biopharm. Drug Dispos. 7,
`327–334.
`Ware, E.C., Lu, D.R., 2004. An automated approach to salt
`selection for new unique trazodone salts. Pharm. Res. 21,
`177–184.
`Wermuth, C.G., 2002. Appendix. In: Stahl, P.H., Wermuth, C.G.
`(Eds.), Handbook of Pharmaceutical Salts: Properties,
`Selection and Use. Wiley-VCH, Winheim, pp. 329–350.
`
`Merck Exhibit 2196, Page 6
`Mylan

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