throbber
This material may be protected by Copyright law (Title 17 U.S. Code)
`
`1
`
`International Journal of Pharmaceutics, 33 (1986) 201-217
`Elsevier
`
`UP 01121
`
`201
`
`Salt selection for basic drugs
`
`Philip L. Gould
`Pharmaceutical Research and Development Department, Pfizer Central Research, Sandwich, Kent (UK)
`
`(Received 24 March 1986)
`(Accepted 30 May 1986)
`
`Key words: Salt form selection — Pharmaceutical salts
`
`
`Summary
`
`An attempt has been made using a Kepner-Tregoe decision analysis approach to provide rationale to salt selection for basic drugs.
`The selection objectives are reviewed in terms of -the ‘essential’ (MUSTS) and ‘desirable’ (WANTS) issues. The desired characteristics
`of the salt form, given sufficient strength and toxicological suitability of the conjugate acid, are then discussed on the basis of the
`various pivotal physicochemical properties; melting point,,aqueous solubility and dissolution rate, stability and _hydrophobicity.
`Several trends are established which can then assist the decision of which range of salt forms to evaluate to overcome a particular
`problem with a basic drug. It is concluded that it is important to view the choice of salt form for development as a compromise, with
`particular focus on the correctly weighted desires to obtain the best balanced choice.
`
`Introduction
`
`Salt formation provides a means of altering the
`physicochemical and resultant biological char-
`acteristics of a drug without modifying its chem-
`ical structure. The importance of choosing the
`‘correct’ salt form of a drug is well outlined in a
`published review (Berge et al., 1977) but, although
`salt form can have a dramatic influence on the
`
`overall properties of a drug, the selection of the
`salt form that exhibits the desired combination
`
`of properties remains a difficult semi-empirical
`choice.
`
`In making the selection of a range of potential
`salts, a chemical process group considers issues on
`the basis of yield, rate and quality of the crystalli-
`sation as well as cost and availability of the con-
`
`jugate acid. The formulation and analytical groups
`are, on the other hand, concerned with the hygro-
`scopicity, stability, solubility and processability
`profile of the salt form, while the drug metabolism
`group is concerned with the pharmacokinetic
`aspects and the safety evaluation group on the
`toxicological effects of chronic and acute dosing
`of the drug and its conjugate acid. Thus, a clear
`comprornise of properties for the salt form is
`required, but the difficulty remains of assessing
`which salt forms are best to screen for a particular
`drug candidate.
`A
`Little,
`if any,
`literature has been devoted (to
`discussing the compromise of properties for salt
`form selection. This review addresses the problem
`of salt form selection for basic drugs.
`'
`
`‘ Correspondence: P.L. Gould, Pharmaceutical Group, Product
`Research and Development Laboratories, Cyanarnid of Great
`Britain Limited, Gosport, Hants, U.K.
`
`Approach to the salt selection process
`
`Walking and Appino (1973) have used the
`Kepner-Tregoe (KT)
`techniques
`(Kepner and
`
`0378-5173/86/$03.50 © 1986 Elsevier Science Publishers B.V. (Biomedical Division)
`
`Mylan Exhibit 1028, Page 1
`
`Mylan Exhibit 1028, Page 1
`
`

`
`TABLE 1
`
`FDA-APPROVED COMMERCIALLY MARKETED SALTS
`
`Anion
`Acetate
`
`Percent “
`1.26 -
`
`Anion
`Iodide
`
`Percent “
`2.02
`
`Benzenesulfonate
`Benzoate
`.
`Bicarbonate
`Bitartrate
`Bromide
`Calcium edetate
`
`‘
`
`0.25
`0.51
`0.13
`0.63
`4.68
`0.25 -
`
`Isothionate i
`Lactate
`Lactobionate
`Malate
`Maleate
`Mandelate
`
`0.88
`0.76
`0.13
`0.13
`3.03
`0.38
`
`Camsylate b ,
`Carbonate
`Chloride
`Citrate
`Dihydrochloride
`Edetate
`‘
`Edisylate °
`Estolate d
`-
`I
`Esylate °
`Fumarate
`
`A“
`
`;
`
`‘
`
`‘
`
`0.25
`0.38
`4.17
`_ 3.03
`0.51
`' 0.25
`0.38
`0.13" 7
`
`0.13
`0.25
`
`i
`
`"V
`
`'
`
`0.18
`Gluceptate I
`~ 0.51
`Gluconate
`0.25
`Glutamate
`Glycollylarsnilate 5 0.13
`Hexylresorcinate
`0.13
`Hydrabamine h
`0.25
`Hydrobromide
`1.90
`Hydrochloride
`42.98
`Hyd.roxynaph-
`thoate
`
`2.02
`0.76
`0.38
`0.88
`0.13
`0.25
`0.64
`1.01‘
`
`0.25
`3.16
`
`Mesylate
`Methylbrornide
`Methylnitrate
`Methylsulfate
`Mucate '
`Napsylate
`. Nitrate
`Pa.mo’a'te" ‘
`(Embonate)
`Pantothenate
`Phosphate/
`' diphosphate
`. Polygalacturonate 0.13
`V Salicylate
`0.88
`Stearate '
`0.25
`Subacetate
`0.38
`Succinate
`0.38
`Sulfate
`7.46
`Tannate
`0.88
`Tartrate
`3.54
`
`'7
`
`_
`
`'
`
`Teoclate J‘
`Triethiodide
`
`0.13
`0.13
`
`0.25
`
`'
`
`
`
`Cation .
`
`Percent “
`
`Cation
`
`Percent 3
`
`Metallic:
`Organic:
`0.66
`Aluminium _
`0.66
`Benzathinek
`10.49
`Calcium _
`‘0.33
`Chloroprocaine‘
`1.64
`Lithium
`0.33
`Choline
`1.31
`Magnesium
`0.98
`Diethanolamine
`10.82
`Potassium
`0.66
`Ethylenedianune
`61.97
`,
`Sodium
`..
`2.29
`Megluminel
`
`
`
`0.66 ZincProcaine 2.95
`
`“ Percent is based on total number of anionic or cationic salts
`
`in use through 1974. b Camphorsulfonate. °1,2-Etha.nedisu.l-
`fonate.
`’d Laurylsulfate.
`"‘ Ethanesulfonate.
`I Glucoheptonate.
`5 p-Glycollamidophenylarsonate.
`h N,N’-Di(dehydroabietyl)
`et_hy1enedia.mine.A i 231-Iydroxyethanesulfonate.
`J 8-Chlorot.h_eo—
`phyllinate. 1‘ N,N’-Dibenzylethylenediamine. 1N-Met.hylgluca-
`mine.
`I
`I
`'
`
`’
`
`Reproduced from .Berge ‘et al. (1977) with permission of the
`copyright owner (.7. Phgrm. Scz'.).
`’
`
`Mylan Exhibit 1028, Page 2
`
`202
`
`Tregoe, 1976) of decision analysis and potential
`problem analysis to aid the selection of a salt
`form. Although their application is more exem-
`plary of the KT method rather than of the specific
`application, the rational process decision analysis
`approach which ‘defines essential and desirable
`attributes as.‘MUSTS’ and ‘WANTS’,
`respec-
`tively, provides a route to initially address the
`problem of,salt form selection.
`
`“G0”/ “N0-G0” issues
`The major “GO”/“NO-GO” (MUSTS) issue
`for salt selection of an ionizable drug is the con-
`sideration of the relative basicity of the drug and
`the relative strength of the conjugate acid. Clearly
`to form a salt the pK,, of the conjugate acid has to I _
`be less than or equal to the pKa of the basic centre
`of the drug.
`.
`A
`.
`.
`Thus the potentialrange of salts of drugs ‘con-
`taining for example triazoyl bases (I; pKa ~ 2) is
`restricted to strong acids (mineral and sulphonic,
`but excluding the carboxylic), whereas imidazole
`bases (II; pKa 6-7) are far less restricted and the
`greatest scope for salt formation occurs for the
`aliphatic tertiary ‘amines (III; pKa 9—10).
`
`‘
`I\}~{\I
`( )[pK.21
`N
`H
`(1)
`
`CH,
`I
`bi \ I
`[pK.61tcH3—N tpK.9i
`N
`I
`CH,
`H
`(H)
`(III) ‘
`
`’
`
`0
`
`The relative acid/base strength of the resultant
`salts also dictates their stability to disproportiona-
`tion, since all salts will be acid and therefore
`potentially reactive towards basic formulation ad-
`ditives..
`__ _ T
`The other essential selection issue for a salt
`
`form is the relative toxicity of the conjugate an-
`ion; some salts clearly fall into a desirable cate-
`gory, some acceptable but
`less desirable ,(both
`“GO”) and some undesirable (“NO GO”). A ta-
`ble of salts used in pharmaceutical products
`marketed in the U.S. up to 1974 is given in Table
`1. It would seem sensible that any acid relating to
`normal metabolism, or present in food and drink
`can be regarded as a suitable candidate for prepar-
`ing salts. Clearly anions that cause irritancy to the
`
`Mylan Exhibit 1028, Page 2
`
`

`
`GI tract should be avoided for some types of drug,
`e.g. anti-inflammatories, laxative surfactant anions
`for anti-secretory drugs and conjugate anions with
`intrinsic toxicity, e.g. oxalatef
`
`Properties desired of the salt form (WANTS)
`Therdesires or ‘WANTS’ of a salt form are
`
`dictated by the nature of the required dosage
`forms,_ their process and desired biological perfor-
`mance. Thus, it is somewhat difficult to provide a
`complete overall specification of ‘WANTS’ for a
`series of salt forms, but
`ideally the bulk salt
`should be completely chemically stable, non—hy-
`groscopic, not cause processing problems, and dis-
`solve quickly from solid‘-’dos‘age forms.
`Because of simple availability
`physiological
`reasons,-the monoproticihydrochlorides have been
`by far the most frequent (f4'40%)‘choice of the
`available anionic salt-forming: species. Thus, there.
`is clear precedent, and an overwhelming argument
`on many grounds to immediately progress to the
`hydrochloride salt -and evaluate other forms only
`if problems with the hydrochloride emerge.
`
`Prepare the hydrochloride; pros andcons
`Kramer and Flynn (1972) suggest that the‘solu-
`bility of an amine hydrochloride generally sets the
`maximum obtainable» concentration _for a given
`amine.
`:
`'
`Many reports (Miyazal<_i et al., 1980,1981) have
`shown that hydrochloride-"shalt formation does not"
`necessarily enhance the solubility "of poorly solu-
`ble basic drugs and result in improved bioavaila-
`bility. This finding is based on the common ion
`effect of chloride on the solubilityproduct equi-
`libriumz
`1
`-
`
`. —
`— Ksp
`BH+C1(s) .= BH;“q + Claq
`
`Hydrochloride salts therefore, have the potential
`to exhibit a reduced dissolution rate in gastric
`fluid because of the abundance of chloride ion
`
`(0.1—O.15 M). Indeed, the Setschenowsalting-out
`constants (k) for chloride are greatest for drugs of
`very low solubility (Fig. 1), and can decrease the
`dissolution rate of the salt to below that of the
`. free base form (Migazaki et al., 1980), which shows
`
`.
`
`' 203
`
`
`
`0.4
`
`0.8
`
`1.2
`
`0.4
`1.6
`LOG S, mglml
`
`0.8
`
`1.2
`
`1.6
`
`Fig. 1. Relationship between solubility in water and saltmlg-out
`constant -at 25°C (left)
`and 37°C (right). Key:
`‘A =
`phenazopyridine; B = cyproheptadine; C = bromhexirie; D =
`trihexyphenidyl; E = isoxsuprine; F = chlortetracycline; G =
`rnethacycline; H = papaverine; and I = demeclocyline.
`
`Adapted from Miyazaki et al. (1981). Reproduced with permis-
`sion of the copyright owner (J. Pharm. Sci.).

`
`that a precipitous drop in drug solubility occurs as
`the free Cl‘ level is increased.
`
`An example of a basic‘ drug showing a strong
`chloride-ion dependence is prazosin.
`1
`‘
`9
`
`N
`
`—N'
`
`N—CO
`
`0-
`
`CH3O
`
`\
`
`cH;o /
`
`Ksp = 2.2X10"6 M @ 30°C
`
`Solubility/mg.ml‘1 @ 30°C A
`
`Base
`Hydrochloride _
`0.1 M HCl ~
`. water water
`0.037 »
`1.40
`0.0083
`
`Chloride, as well as other inorganic anions have
`the potential to form insoluble complex salts with
`weak bases (Dittert at al., 1964), which are then
`potentially less bioavailable than the _free base
`form. The formation of these complex salts is
`controlled by their stability constant Kc.
`
`‘
`Kc
`’
`-J.‘
`‘
`Drug(S) ‘——* Drug(aq) + xH+ : Drug - H: (aq)
`
`.
`
`Mylan Exhibit 1028,’ Page 3
`
`Mylan Exhibit 1028, Page 3
`
`

`
`
`
`204
`
`Evaluation of KC for triamterene (Tr) yields val-
`ues of x=O.5 for. chloride, suggesting that one
`proton solubilizes two molecules of the drug’, i.e.
`the complex is Tr2H+Cl.
`With hydrochloride salts there is frequently an
`‘overkill’ on acid strength, which leads to a very
`low pH for-an aqueous solution (Nudelman et al.,
`1974) of the salt. This can then limit the utility of
`hydrochloride salts in "certain parenteral dosage
`forms, or lead to packaging incompatibilities with
`pharmaceutical metal containers (aerosols).
`Other problems frequently arise as the result of
`the polar nature of hydrochloride salts. Their high
`hydrophilic nature, favouring wettability probably
`as a result of
`the polar ionized groups being
`exposed on the crystal -surfaces,
`leads to. water
`vapour sorption (hygroscopicity) which on occa-
`sions, (may be excessive. This can result
`in
`processing difficulties_(e.g. powder flow) and re-
`, duce the stability of a hydrolytically unstable drug.
`—.This latter effect is exacerbated by the resulting
`very low pH of the loosely bound moisture.
`These-problems can be particularly acute with
`dihydrochlorides.(or disulphates). Also,
`the dif-
`ference in the strength" of the basic centres in
`dihydrochloride salts can lead to a gradual loss of
`one of the hydrochloride moieties by release of
`hydrogen chloride gas (Lin et al., 1972) at elevated
`temperatures or under
`reduced pressure (i.e.
`freeze-drying). Also,
`their extreme polar nature
`results in excessive hydroscopicity (Boatman and
`. Johnson, 1981). eventually leading to deliques-
`cence. .
`.
`.
`
`Thus, progression of a hydrochloride salt should
`be a first move, but if the problems with that salt
`form arises due to some of the reasons outlined,
`then the real selection issue for a salt
`form
`
`emerges-——what trends are available for guidance?
`
`‘ The pivotal issues for salt selection
`
`Each drug and its associated range of dosage
`forms _ will present different salt
`form require-
`ments, and it is perhaps best to discusssalt selec-
`tion further byloutlining some of the trends ‘in salt
`properties that may facilitate selection.
`
`The pivot of melting point
`
`A change in the development of a compound
`from the free base to a salt may be promoted by a
`need to moderate the kinetics and extent of drug
`absorption, or to modify drug processing. Unfor-
`tunately these desires may be mutually exclusive,
`as the balance between these properties is fre-
`quently pivoted around the melting point of the
`salt form. For example, an increase in melting
`point is usually accompanied by a reduction in
`salt solubility (the ideal solubility of a drug in all
`solvents decreases by an order of magnitude on an
`increase of 100°C in its melting point), whereas
`high melting crystalline salts are potentially easier
`toprocess...
`_
`..
`.
`The increase or decrease in_ melting point of a
`series of salts is usually dependent on the control-
`ling effect of crystallinity from the conjugate an-
`ion. This isgexemplified by considering an experi-
`mental drug candidate (UK47880) which has a
`basic pKa of T 8, and therefore gives access to a
`wide variety of salt forms:
`'
`
`
`
`‘Sf:
`
`UK-47880
`
`melting point, 74°C
`
`Salts prepared from planar, high melting aromatic
`sulphonic or hydroxycarboxylic acids yielded
`crystalline salts of correspondingly high melting
`point
`(see Table 2), whereas flexible aliphatic
`strong acids such as citric and dodecyl benzene
`sulphonic yielded oils. Thus,
`the comparative
`planar symmetry of the conjugate acid appears
`important for the maintenance of high crystal
`lattice forces. This is shown by the melting point
`of the conjugate acid being highly correlated with
`. the melting point of the‘resultant salt form (Fig.
`2). Therefore the highly crystalline salts are in this
`case best suited to reducing drug solubility.
`Alternatively it should also be feasible to build
`up crystal lattice forces of drugs with good hydro-
`gen bonding potential, by considering syrnmetry
`and hydrogen bonding potential of the conjugate
`acid. One salt
`series of
`interest-
`is
`that
`for
`
`Mylan Exhibit 1028, Page 4
`
`Mylan Exhibit 1028, Page 4
`
`

`
`TABLE 2
`
`MELTING POINT OF SALTS 01: EXPERIMENTAL COM-
`POUND (UK47880) AND THE CORRESPONDING =coN—
`IUGATE ACID
`
`Melting point (°C) Legend
`Salt
`Conjugate
`F13‘ 2
`acid
`
`
`.
`
`74
`
`235
`
`280
`
`170
`156
`
`223
`
`190
`158
`
`220
`
`4
`
`_ A
`G
`
`D
`C
`_
`E
`
`UK 47880’; free base
`pamoate (embonate)
`4—hydroxynaphthalene-
`1-sulphonate
`-
`Salicylate 1
`3—hydroxynaphthalene-
`2'-caxboxylate
`.
`2—hydroxynaphthalene—1
`B
`120
`145
`—ca.rboxylate
`'
`“_
`'
`F
`225
`2,34
`anthraquinone-3-sulphonate
`—
`‘—
`-
`20
`dodecylbenzene sulphonate
`—
`~ 20
`.
`113'
`mesylate
`'
`T
`
`
`
`. ‘ 20 153citrate -
`
`205
`
`3GQQ
`
`60
`
`80
`
`300
`200
`100‘
`MELTING POINT AClDl°C
`
`
`
`
`
`
`
`MELTINGPOINTSALT/t“c
`
`Fig. 2. Plot of melting point of UK47880 salts vs melting point
`of conjugate acids. Legend given in Table 2.
`
`A
`
`7
`
`N
`H
`
`OH
`I
`
`C
`
`' CF3 »
`
`
`
`
`
`
`
`epinephrine
`
`Ho—cHcH2NHcH;
`
`I
`OH
`
`OH __;'._
`
`Salt form
`
`epinephrine
`ta.r’r_rate
`
`maleate
`malate
`fumarate
`
`'
`
`Melting
`point,(°C)
`
`157 ‘
`149
`
`182 -_
`170 A
`103
`
`_
`
`p
`
`hydrogen bonding acids such
`where small
`as malonic andvmaleic gave higher melting salts,
`whereas the larger bitartrate and presumably.sym-
`metrically unfavoured fumarate. gave salts of lower
`melting point.
`‘
`
`Salt form
`
`m.p; salt m.p. acid
`
`solubility
`
`.
`\
`
`, M
`Free base
`HCl
`.
`DL-lactate
`L—lactate
`2-hydroxyethane
`sulphonate
`Mesylate
`. Sulphate
`
`/
`
`_
`
`<°c)
`215
`331
`172
`192
`
`. 251
`290
`270 -
`
`i
`
`<°C)
`
`17
`5'3
`
`.
`
`'
`
`‘
`
`(mg/ml)
`.7.5
`13‘
`, 1850
`925»
`
`'
`
`620
`300
`20
`
`.
`
`The relationship between aqueous’ solubility
`",(SW) and melting point is shown diagrammatically —
`.
`in Fig.» 3, where log1SW is correlated overa range
`. of salts with the inverse of the melting point.
`Interestingly with this compound, the solubility of
`the hydrochloride salt-in water is only approxi-
`mately twice that of the free base, whereas the low
`«melting DL-lactate provides a 200-fold advantage
`over the free base in terms of solubility, which is "a
`result in part of the reduced lattice energy.
`
`.

`Melting point and aqueousis0lub'z"Zz'ty~ V
`The trends in melting point“ (m.p.) andaqueous
`solubility alluded to above are exemplified in the-
`salts of a high melting antimalarial drug (Aghar-
`kar et al., 1976).
`.
`'
`‘
`
`1Melting point and chemical stability '
`The stability of .organic compounds in the solid
`state-is intimately related to the strength of the
`‘crystal lattice. Since the forces between molecules
`: in a crystal are small compared with the energy.
`
`Mylan Exhibit 1028, Page 5
`
`
`
`Mylan Exhibit 1028, Page 5
`
`

`
`
`
`206
`
`essentially controls the formation and extent of
`eutectic melts.
`
`As an additional aspectto the strength of crystal
`forces, the balance of the amorphous to crystalline
`nature in solid salts» can dramatically affect their
`stability. This is exemplified by the sodium salts
`of ethacrynic acid (Yarwood et al., 1983).
`
`V
`
`V
`
`m.p. (9 C) ‘
`' % remaining after
`9 days @ 60° C
`
`Sodium ethacrynate
`
`Crystalline
`
`’ Amorphous
`
`200
`
`100
`
`'
`'
`
`_
`
`92
`
`These results are consistent with the concept of an
`amorphous material being a highly viscous con-
`centrated solution and show that
`the stronger
`crystalline. lattice forces result
`in superior solid
`state stability.
`''
`A
`
`Melting point and formulation processing
`Salt formation is frequently employed to raise
`the melting point (and crystallinity) of the drug
`species being processed. However, published work
`concerning this type of manipulation is somewhat
`sparse.
`,
`A
`
`The melting point of _drug salts can dra.mati-
`cally affect their physical storage. Drugs (or salts)
`with low melting points generally exhibit plastic
`deformation (Jones, 1979) and thus during storage
`the stress exerted by the bulk mass on the asperity
`points o_f interparticulate contact can lead to the
`formation of localized welds leading to bulk ag-
`-gregation. Also, if the sublimation temperature is
`low (e.g. ibuprofen, m.p. 76°C), intraparticulate
`voids can be bridged by sublimed drug again
`leading to aggregation. Thus on storage, the bulk
`drug salt will begin to cake and aggregate, thereby
`altering significantly its flow, compression and
`long-term dissolution properties.
`Melting point also has a crucial role in drug
`processing, in particular comminution and tablet-
`ing. Since low melting compounds tend to be
`plastic, rather thanbrittle, they comminute poorly,
`and frictional heating causes melting and deposi-
`tion of the drug on the screens and pins of the mill _
`causing it
`to ‘blind’. For production ‘of
`fine
`pharmaceutical powders this aspect is crucial to
`judging the correct level of filler to allow efficient
`
`Mylan Exhibit 1028, Page 6
`
`I
`
`
`
`SOLUBILITYIgm!“
`
`P I
`
`
`
`0.01 m_imm__lm_____1m_1__
`1.5
`1.3
`.
`2.0
`2.2
`
`[1/MELTING POINT) x
`
`1a3r<"
`
`< Fig. 3."Plo‘t of aqueous solubility vs inverse of absolute melting
`point for a series of salts of a hydrophobic antimalarial drug.
`Data taken from Agharkar et al. (1976).
`'
`A
`
`required to break chemical bonds, liquefaction of
`the solid (and an increased frequency of molecular
`collisions) occurs before degradation begins. Thus
`the melting point of a compound can be an‘ im-
`8 portant factor in determining stability.
`.
`) Degradation of solid drugs, when it is observed,
`usually occurs in the surface film phase and is
`accompariiecl bylthe formation of a liquid phase at
`temperatures below the normal meltingpoint of
`the solid. Using this so-called ‘liquid layer’ ap-
`proach, Guillory and Higuchi (1962) investigated
`the stability of esters of vitamin A employing the
`following equation to determine the relationship
`between degradation rate and melting point.
`
`log K=
`
`AAH[1}_AH
`
`where Tm = normal melting point; Td = depressed
`storage temp.‘ = storage temperature; K = degrad-
`ation rate constant; AH = heat of fusion.
`Thus, for a series of related compounds subject
`to a storage temperature Td, the logarithm of the
`degradation rate constant is inversely related to
`the absolute melting point of
`the compounds.
`Although this approach may be somewhat simplis-
`tic it may have utility as a method of assessing the
`bulk stability of ~non—hygroscopic salt forms.
`The melting point of a salt form also has some
`influence on its relative compatibility with drug
`combinations (Hirsch et'al., 1978) or formulation
`, excipients (Li Wan Po and ‘Mroso, 1984) since it
`
`Mesylate
`Edisylate
`L‘ Lactate
`DL-Lactate
`
`‘
`
`
`
`Mylan Exhibit 1028, Page 6
`
`

`
`manufacture using a cost-effective feed rate.
`Salt melting point can also have important
`implications for particle bonding on compression
`for tableting. Since bonding on compression oc-
`curs by point welding at the deformed or frag-
`mented particle surfaces, then at a fixed tempera-
`ture and pressure, a lower melting species would
`be expected to improve bonding. However,
`the
`pressure on the powder (and the eutectics formed
`with the other excipients) suppress the melting
`point further. The Skotnicky equation defines the
`fall in melting point (Tm) with the pressure on the
`solid (PS)
`A
`
`dTm
`dPs'
`
`~VST
`Hf
`
`where AHf = heat of fusion; V5 = volume of solid;
`T= temperature, and therefore as well as those
`salts which are intrinsically low melting, salts of
`different values _of AHf would be expected to have
`different abilities to cold weld in the compression
`process. If we compare, for example, the melting
`points and heats of fusion of the salts of an
`experimental drug candidate:
`
`.
`
`.
`
`—
`
`Salt_
`Hydrochloride
`Mesylate
`Tartrate
`Citrate
`Phosphate
`Acetate
`
`.
`
`'
`
`Tm (°c)
`280
`135
`213
`180 s
`2'50 1
`180
`
`,
`
`‘
`
`AHf (kJ-mol‘1)
`56.5
`V »20.5'
`63.6
`27.2
`136.5 -
`- 167.9
`
`‘
`
`-I
`
`r
`
`the data suggest that the low melting point and
`low AHf for the mesylate_ salt would make it the
`most suitable candidate, on bonding "grounds, for
`a direct compression tablet. Since the melting
`points of compounds are reduced under pressure,
`the solubility of salt forms would be expected to
`increase with increasing pressure. This can p_oten.-
`tially cause the formation of solutions of the salts
`in the film of absorbed moisture on the surface of
`the drug (and excipient) particles which then may
`have an effect on drug bonding (Parrot, 1982) or
`cause‘ the drug to adhere to the punches on com-
`pression (Wells and Davison, -1985).
`-
`
`Conclusion
`
`_
`
`The consideration of melting point. is a key
`
`207
`
`parameter in assessing the ‘viability’ of certain
`salt forms. In general, an increase in melting point,
`usually by maximizing or encouraging crystal sym-
`metry, leads to reduced solubility in all solvents,
`but generally improved stability, particularly if
`salt formation results in a crystalline solid, and
`easier formulation processing. For a specific salt
`form for parenteral use, i.e. where solubility and
`resultant pH is a major issue, a low melting point
`salt produced using a soluble fairly weak acid (see
`next section) probably made in situ is likely to be
`preferable.
`
`The pivot of drug solubility
`
`A There are various solubility issues that can de-
`cide the viability of a particular salt form and it is
`perhaps worth addressing these separately to iden-
`tify trends that may aid salt selection.
`
`Aqueous solubility per se
`As indicated earlier, the solubility of a drug can
`enhanced dramatically by salt;
`formation
`be
`(Agharkar et al., 1976). This enhancement may
`arise from‘ a reduction in melting point, or from
`improved water-drug interactions. A good exam-
`ple of this is with the salts of chlorhexidine (Senior,
`1973), where increased water solubility" was not’
`‘only produced by a lowering of melting point, but
`by increasing‘ the hydroxylation of the conjugate
`acid.
`'
`'
`'
`
`Chlorhexidine
`
`Cl@NH— pNHpNH(cH2)6NHfiNHfiNH@c1
`
`|
`NH NH
`
`NH NH
`
`— Salt 9
`
`Stnlcture
`
`above
`_
`.
`base
`dihydrochloride HCl
`
`_
`
`_
`
`_
`
`p
`
`,
`
`di-2hydroxy-
`
`naphthoate-
`
`co2H
`
`|
`
`.OH —
`
`_
`
`~
`
`I
`
`Melting Solubility
`point
`% W/v
`(° C)
`@ 20 ° C
`
`134
`261
`
`0,008
`(0.06
`
`—
`
`V 0.014
`
`diacetate .
`dilactate
`digluconate
`
`_
`
`154
`CH3CO2H
`—
`CH3CHOHCO2H
`HO2C(CHOH)4CO2H low
`
`1.8
`1.0
`70‘
`
`Mylan Exhibit 1028, Page 7
`
`Mylan Exhibit 1028, Page 7
`
`

`
`
`
`208
`
`-
`
`.
`
`The above data exemplify the importance of
`considering the hydrop_hilic nature of the con-
`jugate anion, as well as its role in controlling
`crystallinity, when considering the potential solu-
`bility of salts.
`.
`Reduced aqueous» solubility may occasionally
`be a crucial development factor for a drug, e.g. for
`an organoleptically acceptable or chemically sta-
`ble suspension. Such systems demand salts of low
`solubility, but recent experience with a series of
`purposely designed insoluble salts of an experi-
`mental drug candidate also highlighted the need to
`consider the solubility and pKa of the conjugate
`anion.
`
`BH4 X15, =BH;;q, +X;q
`B - free base of
`salt
`1LK,, K, 5lL+H*'
`experimental drug
`B<s> *‘—‘ B<aq>
`HX<aq> *‘—‘ H799
`H+
`
`-111C aoove 1OI11C eq111l.lDI‘1a snow that even sparing
`solubility of the salt means that the level of the
`conjugate anion in solution will depend markedly
`“ on the pH of the fluid. Consideration of the above
`for a pamoate salt, which has pKa’s of the parent
`acid of 2.5 and 3.1 and virtually insoluble un-
`ionized form, indicates that solutions of pH 5-6
`will drive the equilibria to the right, with full
`precipitation of the free acid HX(S) and liberation
`of a full component in solution of the ionized base
`(BH+). However, if we consider the hydroxynaph-
`thalene sulphonic acid (pKa = 0.11) then this sys-
`tem provides ‘insolubility’pover a much wider pH
`range and is therefore far more tolerant to fluctua-
`tions in the fluid pH.
`The ‘above aspect is important when consider- '
`ing the potential use of
`‘insoluble’
`salts (e.g.
`pamoate)
`to control
`the absorption of a drug
`candidate..For example,
`the in vitro dissolution
`rates of the dimaleate and pamoate salts of a drug
`candidate were compared in simulated gastric and
`-intestinal fluid. The dissolution rates were essen-
`tially identical
`in the former fluid, with rapid
`deposition of the pamoic acid and liberation of
`the free base, whereas in the latter the pamoate
`salt exhibited a much slower dissolution rate than
`the maleate. Therefore ‘control’ on the drug ab-
`sorption (and toxicity) may then depend -on the
`duration of gastric residence and the pH of the
`gastric contents. Thus aspects such as food vs the
`
`fasted state are also important. In fact in this case,
`the bioavailability in the dog of the two salt forms
`when dosed orally from ‘a standard capsule formu-
`lation were of the same order; 24% for the pamoate
`and 39% for the maleate.
`'
`
`is the v- dissolution rate of a drug
`Usually it
`which is of major importance to the formulation
`and as a rule a salt exhibits a higher dissolution
`rate than the base at an equal pH, even though
`theyhave the same equilibrium solubility. This
`latter effect, which is exemplified by theophylline
`salts (Nelson, 1957), is due to the salt effectively
`acting as its own buffer to alter the pH of the
`diffusion boundary layer,
`thereby increasing the
`apparent solubility of the parent drug in that
`layer. Thus, administration of basic drugs as their
`salt forms (e.g. tetracycline hydrochloride) ensures
`that stomach emptying rather than in vivo dissolu-
`tion will be" the rate-limiting factor in their absorp-
`tion. It is also possible that increased drug absorp-
`tion may occur with salts due to their effect on the
`surface tension ofithegastrointesftinal fluids (Berge
`et al., 1977).
`
`Salt solubility and pH of salt solutions
`__ Enhancement of the aqueous solubility of a
`drug by salt formation can occur due to_ dif-
`ferences in the pH of the saturated salt solutions.
`A soluble acid salt of a weakly basic drug will
`cause the pH to drop as the salt is added to the
`solution. This pH drop will, in turn cause more
`drug to dissolve, and this process will continue
`until the pH of maximum solubility is reached (see
`Fig. 4). The equilibrium solubility(ies) are then
`given by:
`
`s = s+(1 + 10PH~PK«)
`
`i.e. when the ionized form is
`for pH=pHmax,
`solubility limiting and
`
`s = s,(1 + 10PK«-PH)
`
`for pH=pHmax, where the unionized form is
`solubility limiting and pHmx is given by the solu-
`tion of the equality of pH for the above two
`
`Mylan Exhibit 1028, Page 8
`
`Mylan Exhibit 1028, Page 8
`
`

`
`SOLUBILITYmglml
`
`Fig. 4. Solubility of A in water at ambient temperature (~
`23° C) as a function of pH. -All data are in mg/ml calculated in
`terms of free base equivalent. The lines drawn through the data
`are theoretical and were calculated using 0.067 mg/ml as the
`free base solubility, 11.5 mg/ml as the hydrochloride solubility
`and 8.85 as the pK,,. Data by both gravimetric (I) and GLC
`(O) procedures were in good agreement.
`
`Adapted from Kramer and Flynn (1972), with permission of
`the copyright owner (J. Pharm. Sci.).
`
`equations where
`
`pHmax = pKa + log
`
`+
`
`1
`
`and implies that both freebase and salt form can
`exist
`simultaneously in equilibrium with the
`saturated solution.
`,
`
`large pH shifts on dissolution of salts
`Thus,
`suggests that a large amountof conjugate acid is
`dissociating and therefore, a relatively high solu~
`bility is- then obtained. If we consider physiologi-
`cal pH, a low pKa fora conjugate acid of high
`aqueous solubility, would appear to give the best
`change of obtaining the lowest pHmax and the
`highest aqueous salt solubility. For example, the
`solubilities of a series of salts of a drug candidate 0
`and the‘ pH of the saturated solutions were as
`follows:
`
`Type
`
`Salt
`
`Conjugate acid
`
`209
`
`pHm,,,,
`
`Solu-
`, Solu-
`bility
`bility
`(mg/
`.
`.
`(mg/
`
`ml)
`ml)
`
`pKa
`
`Hydro-
`chloride
`Mesylate
`Tartrate
`Citrate
`Phos—
`
`2.71
`2.57
`4.21
`3.30
`~
`
`35.9
`51.2
`0.49
`2.16
`
`— 6.1
`— 1.2
`3.03 _
`3.13
`
`1470 ‘
`2400 ,
`
`2.15
`10.31
`5.31
`phate
`
`
`
`5.29 8.04Acetate 4.76
`
`indicating that the salts of stronger acids (HCl,
`methanesulphonic) produce the lowest slurry pH
`and the highest salt solubility. In this case the
`solubility and resultant low pH of the hydrochlo-
`ride is suppressed by the common ion effect. The
`solubility of salts such as the lactate (pKa of
`conjugate acid is 3.85, with infinite solubility) may
`offer significant advantage over for example the
`acetate, tartrate and citrate.
`'
`-
`The pH of. a salt solution can‘ be a deciding
`factor in the selection of a salt for aparenteral
`dosage form. Ideally to avoid pain on injection the
`pH of i.v. parenterals should be between pH 3 and .
`9, and so highly acidic salts such as the hydrochlo-
`ride and mesylate are probably best replaced by
`an acetate salt.
`.
`'
`
`Kramer and Flynn (1972) have shown that for
`a series of hydrochloride salts, by making analysis
`of the differential heat of solutions of the ionized.
`
`that the temperature de-
`and unionized species,
`pendencies of the solubilities of the hydrochloride
`salts were considerably lower than those of the
`corresponding free base form. This may have im-
`portant implications for solution dosage form des-
`ign and storage conditions.
`
`‘A
`Salt solubility and salt stability
`As well as the relationships between salt melt-
`ing point and stabilityraised earlier, it is -also clear
`that
`low solubility and low hygroscopicity can
`contribute significantly to the stability of a salt
`form. The former aspect is obviously important in
`developing a stable aqueous suspension formula-
`tion of a hydrolytically unstable water soluble
`<;
`
`Mylan Exhibit 1028, Page 9
`
`Mylan Exhibit 1028, Page 9
`
`

`
`210
`
`drug; e.g. penicillin-benzithine.
`For salts of weak bases, the moisture associated
`with the bulk can be very acidic (the salt will
`
`buffer the available moisture), and can potentially
`cause severe hydrolytic degradation of the parent
`drug. Classic examples of this phenomenon are
`thiamine salts (Yamarnoto et al., 1956, 1957) where
`the stability is related to their hygroscopicity,’
`aqueous solubility and the resulting pH.
`,
`Thus, to improve drug stability by salt forma-
`tion,
`it is clearly not only important to control
`hygroscopicity, but also to consider carefully the
`strength‘ of the conjugate acid used to form the
`salt. This is particularly important for compacted
`dosage forms where salt. and excipient share the
`available moisture, particiularly when the majority
`of the available moisture comes from the excipient
`rather than the drug. Thus, assessment of salt
`stability in compressed and —non-compressed sys-
`tems is «an important activity inpreformulation
`studies. However,
`in selection terms
`salts of
`mineral acids will produce a lower pH, and higher
`solubility in the available moistureand therefore
`produce a more hostile stability environment than
`that from a sulphonate or carboxylate type salt. It
`is also

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