throbber
(cid:11)(cid:12)(cid:14)(cid:13)(cid:12)(cid:15)(cid:16)(cid:4)(cid:10)(cid:15)(cid:5)(cid:4)(cid:7)(cid:6)(cid:8)(cid:9)(cid:4)
`(cid:12)(cid:13)(cid:14)(cid:4)(cid:13)(cid:17)(cid:21)(cid:18)(cid:21)(cid:18)(cid:20)(cid:19)(cid:4)(cid:6)(cid:4)(cid:16)(cid:15)(cid:13)(cid:4)(cid:9)(cid:5)(cid:8)(cid:7)(cid:9)(cid:5)(cid:11)(cid:10)(cid:11)(cid:4)
`
`Apotex Exhibit 1012.001
`
`

`

`Pharmacy Library
`University of Wisconsin - Madison
`2130 Chamberlin Hall
`425 N. Charter Street
`Madison,WI 53706-1608
`
`Library of Congress Cataloging in Publication Data
`Main entry under title:
`
`Encyclopedia of Pharmaceutical Technology.
`editors: James Swarbrick, James C. Boylan.
`
`Includes index.
`
`1. Pharmaceutical technology—Dictionaries.
`II. Boylan, James C.
`2. Drugs—
`[DNLM: 1. Chemistry, Pharmaceutical-encyclopedias.
`encyclopedias.
`3. Technology, Pharmaceutical—encyclopedias. QV 13 E565].
`RSI92.E531988 615’.1’0321-dc19
`
`I. Swarbrick, James.
`
`COPYRIGHT © 1996 BY MARCEL DEKKER, INC. ALL RIGHTS RESERVED.
`
`Neither this book nor any part may be reproduced or transmitted in any form or by any
`means, electronic or mechanical, including photocopying, microfilming, and recording,
`or by any information storage and retrieval system, without permission in writing from
`the publisher.
`
`MARCEL DEKKER, INC.
`270- Madison Avenue, New York, New York 10016
`
`LIBRARY OF CONGRESS CATALOG CARD NUMBER 88-25664
`ISBN: 0—8247—2812—2
`
`Current printing (last digit):
`10
`9
`8
`7
`6
`5
`4
`3
`
`2
`
`1
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`Apotex Exhibit 1012.002
`
`Apotex Exhibit 1012.002
`
`

`

` This material may be protected by Copyright law {Title ”l"? US. Code}
`
`Salt Forms of Drugs and Absorption
`
`Introduction
`
`Salt formation is frequently performed on weak acidic or basic drugs because it is a
`relatively simple chemical manipulation which may alter the physicochemical, formu-
`lation, biopharmaceutical, and therapeutic properties of a drug without modifying the
`basic chemical structure. Salt selection has been largely semi-empirical, based on con-
`sideration of cost of raw materials, yield, ease of preparation and purification, etc. Al—
`though attempts have been made to apply “decision analysis” and “potential problem
`analysis” to select salts and help predict salt performance [1], the choice of which salt
`to use remains a difficult decision.
`
`The ideal characteristics of a salt are that it is chemically stable, not hygroscopic,
`presents no processing problems, dissolves quickly from solid dosage forms (unless it
`is formed with the intent to delay dissolution), and exhibits good bioavailability.
`The literature contains a large amount of information on salts; however, much of
`the early research addresses the use of salt formation to prolong the release of the active
`component, thereby eliminating various undesirable drug properties[2—6]. This article
`supplements an extensive review published in 1977 [7], providing a literature overview
`of approximately 40—45 years. Its objectives are to present potentially useful salts, their
`effect on the properties of the parent drug, and a decision tree for choosing the most
`desirable salt form(s) for development.
`
`Potentially Useful Salts
`
`Salt formation is one of the simplest chemical reactions, involving either a proton transfer
`or a neutralization reaction between an acid and a base. The relative strength of the acid
`or base, or the acidity and basicity constants of the species involved, significantly in-
`fluences the occurrence of the reaction and provides a measure of the stability of the
`resulting salt. Theoretically, every compound possessing acidic and/0r basic properties
`can participate in salt formation.
`Salt forms that have been clinically evaluated in humans or were commercially
`marketed through 1993 are shown in Tables 1 and 2, compiled from the drug mono—
`graphs listed in Martindale, The Extra Pharmacopoeia, 30th'ed. [8]. Table 1 gives all
`anionic salt forms, Table 2 all cationic forms. The relative frequency (as a percentage)
`of use for each salt type was calculated based on the total number of anionic or cat~
`ionic salts used through 1993.
`The monoprotic hydrochlorides are by far the most frequent choice of an anionic
`salt—forming radical, probably for physiological reasons and simple availability. For simi—
`lar reasons, sodium is the most predominate cation. These findings are identical to those
`reported in a‘similar survey [7] from 1977, even though they are based on twice the
`number of salts as the earlier study. Other comparisons between this and the previous
`review show an increase of approximately 40% in the types of anionic salts and approxi—
`453
`
`Apotex Exhibit 1012.003
`
`Apotex Exhibit 1012.003
`
`

`

`454
`
`Salt Forms of Drugs and Absorption
`
`
`
`
`
`$50qunow:“ESSAdoE<“ESSAneg
`
`
`
`
`
`
`
`
`
`
`
`
`33Ebfiobno250m3%Eofidoowqdmnm0803Fm..m<._.
`
`
`
`
`
`2483:2no.0Muumwmom.mmomuwflonnmoomNS.0Sandbagno.0Baconno.0Bmfifiwoodm
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`0N083802.088082000.N8884»m.0885No.0Bananano.003389033884
`
`
`
`
`
`3.0«$55N9083836no.0ouwflwfimmwflboo/w,
`
`333338360
`
`
`
`AflwnmmonmfiozoAflfioumofiznamomfinnv
`
`
`
`
`
`nmAABwqonfimooumoammno.0038330ma.0Bfimfi<2.0oufiflwomoS.0898535no.0owmtmmmflboomw
`
`
`
`
`
`
`
`
`
`
`
`no.0Emmofiofiwm«moouwnmmonmeoobw9.0oumfinoamoflg
`
`
`
`
`
`
`
`no.0835$Q.083839no.0Bmfiooqflnfioaoéng
`
`
`
`
`
`
`
`
`
`m;Baasgafioafifi8.0$332385268850?
`
`A8288Magnonmowfiazoobw-3
`
`
`
`
`
`no.03305no.0885005omomagnummi3.39858.0afifiafiwmmoeugmé
`
`
`
`
`
`
`
`no.0fimbmom2.0Bacafimflfiom0N0owflnmom
`
`
`
`
`
`
`
`mm.0ammuumwoomno.0BwBBOmmhbamn0.0BEEP..wowm
`
`
`
`
`
`
`
`
`
`Q2560BowoflbowfiozvAmwmnoflsmoqflfimo
`
`
`
`no.0oumquBoflwwEom0N.0“wagonnmQ.08%,Em
`
`
`
`
`
`ON.00:380onnm.HoEEoSoénmmm.098535
`
`BmoaonQnNmoaxoénm-$é
`
`
`
`m338%235258.9.vuuoaooimone288
`
`
`
`
`
`
`
`
`
`omocowgnoommno.0oEwoBHunmand03805
`
`
`
`
`
`
`
`
`
`wno835mgn0.ouumuofinmoqogoemxoénmno.0owflEoHBbwm
`
`
`
`
`
`
`
`
`
`0N033meno.0Smoflsnbmoénm8.0832385
`
`
`
`
`
`no.0Smbnflbmoumno.0oumoflammnnxohvnmmmoBflnwawo
`
`
`
`
`
`
`
`Auumqofidmuonmamov
`
`Apotex Exhibit 1012.004
`
`Apotex Exhibit 1012.004
`
`

`

`oHeauafiogeoaoé
`
`Fmmopwnofinmmmmfionxoénm-@HNmo.SmEoonmNmoowwnoHfiomHmm.moHioEU
`
`
`
`
`
`Bfioogxonoameofiusno.ouHmHnoHmeoodnm‘no.o055580waHw.NMambammm.m051%mm.o880meno.o
`
`
`
`
`
`
`
`manno.oBfimumapm‘HH.HMUEHEHow.ooumnofimo
`
`amouwuobdmomwflofim-vénmasoobnegamfiemmod@8thgo833%no.oBNHHmoSoHOD.Amwmqofldmoswnunohofloév
`
`
`
`
`
`cm‘mwo8%anmooQEQA‘Hno.o83320
`
`
`
`.mno.oUBNHHmonoH_vH.m03234mH.oBmENHonU
`
`
`
`
`
`.m.mmd.3208ercam3%on>3QESEUBHUEQA8.0emaafiefim3833%:8.00338.633
`
`
`
`
`
`
`
`.mno.o8883anom.ouoHHVoHHbtoSHno.o8&0mem.8.0BNQEOEHoweoflaosaaozno.oamnesia
`
`
`
`nHSNEMSHSHE$882535.
`
`
`
`
`
`
`
`:3Baggagem3Bataan:8dBawam
`
`
`
`omoBarmanwooBflHdenfioSHmH.oBEBE
`
`
`
`ABNHNANOnpmoééqmigmflbAoomdomHsmEonwfim-mhHV
`
`
`
`
`
`Amumnobnw-v-oaflobAoHflHdmH3310
`
`
`
`
`
`
`
`no.oDueseugno.oaaofimoflonoz2.0033mm
`
`mH.oupmoqmooombno.o88:2no.o.oEBoSHnfim
`
`
`
`So385%m3Bameamzno.ooataaam
`
`
`
`GUNomnmooaoAmumnobnmoqwflmv
`
`
`
`ABNOHHEHHNQHQOHHVEHVomo@fimobnmkuEHwfiHHmfl/HfifiHoBarmanZno.oQRONHHVHHunH
`
`
`
`
`
`
`
` .898359335anqmnmnxoénmv
`
`
`
`
`
`.mmmHammobwmm:3£3oEoEm[Ho5835H88noH623wH5080mm
`
`
`
`.5388Babomoooumwnmafifiolunfimm@3863“@8383?
`
`
`
`
`
`455
`
`Apotex Exhibit 1012.005
`
`
`
`
`
`.Sufismundocumnbfiudmowflob"N-HnfioEE-mhHouonnmboné"Wm“Ho
`
`Apotex Exhibit 1012.005
`
`

`

`456
`
`Salt Forms of Drugs and Absorption
`
`TABLE 2 Cationic Pharmaceutical Salt Forms Currently in Use
`
` Organic Cation Percenta Metallic Cation Percenta
`
`
`
`
`
`
`
`Aluminum
`Bismuth
`
`Calcium
`
`Lithium
`
`Magnesium
`Neodymium
`Potassium
`
`Rubidium
`Sodium
`Strontium
`Zinc
`
`1 .35
`0.30
`
`12. 18
`
`0.90
`
`4.51
`0. 15
`9.77
`
`0.15
`57.74
`0.30
`1 .05
`
`,7
`
`1 .95
`O. 15
`
`0.45
`
`0.15
`
`O. 15
`0. 15
`0. 15
`
`0.60
`0. 15
`0.45
`0.60
`0. 15
`0. 15
`
`0. 15
`
`0. 15
`0. 15
`
`0. 15
`
`0. 15
`0.30
`2.41
`
`p
`
`.
`
`Ammonium
`Benethamine
`(N—Benzylphenethylamine)
`Benzathine
`(N, N’ —Dibenzylethylenediamine)
`Betaine
`((Carboxymethyl)trimethylammonium hydroxide)
`Carnitine

`Clemizoleb
`Chlorcyclizine
`1—(4—Chorobenzyhydryl)—4—methylpiperazine)
`Choline
`Dibenzylamine
`Diethanolamine
`Diethylamine
`Diethylammonium
`Eglumine
`(N—Ethylglucamine)
`Erbumine
`(t—Butylamine)
`Ethylencdiamine
`Heptaminol
`(6~Amino—2—methylheptan—'2—ol)
`Hydrabamine
`(N, N' —Di(dihydroabietyl)ethylenediamine
`Hydroxyethylpyrrolidone
`Imidazole
`Meglumine
`(N-Methylglucamine)
`0.45
`Olamine
`0.90
`Piperazine
`0.5 1
`4—Phenylcyclohexylamine
`0. 15
`Procaine
`0. 15
`Pyridoxine
`O. 15
`Triethanolamine
`0.90
`Tromethamine ‘
`(Tris(hydroxymethyl)aminomethane)
`
`“Percent based 011 total number of cationic salts in use through 1993.
`b1~p—Chlorobenzyl—Z-pyrrolidin—1’ —y1methylbenzimidazole
`
`mately 80% in the types of cationic salts in use. This may be indicative of a trend to
`modify or optimize the properties of a substance through salt formation as opposed to
`more complex molecular modifications. In addition, the interest in polymer—drug salts
`for controlling drug release is indicated by the appearance of polistirex and policrilix
`salts.
`
`It is well documented that due to differences in physical, chemical, and thermody—
`namic properties imparted by the salt—forming species, various salts of the same com-
`
`Apotex Exhibit 1012.006
`
`Apotex Exhibit 1012.006
`
`

`

`Salt Forms of Drugs and Absorption
`
`457
`
`pound often behave differently. Knowledge that a particular salt form imparts enhanced
`water solubility, reduced toxicity, or slow dissolution rate to a drug molecule greatly
`benefits chemists and formulators. Although some generalizations such as the statement
`by Miller and Heller on water solubility associated with carboxylic acid salts [9] can
`be followed, drug use and history frequently dictate the salt form selected. For example,
`most contrast agents requiring water solubility are meglumine salts, whereas many of
`the newer therapeutic peptides (i.e., buserelin, nafarelin, octreotide) are acetates. Many
`of the antibiotics administered intravenously are sodium salts. This indicates that the
`drug class, history of use and local tolerance, and possibly regulatory acceptability
`influence the selection of the salt form.
`Both pamoic acid and alginic acid have been shown to prolong action by forming
`slightly soluble salts with certain basic drugs. The incorporation of pamoate salts in
`sustained-release preparations has been reviewed by Saias et a1. [10]; numerous examples
`can be found in the literature [11—16]. Alginic acid salts of streptomycin [17] and pilo-
`carpine [18] have been prepared and shown to provide sustained action.
`A unique way of prolonging action through salt formation was demonstrated by
`Malek and co—Workers [l9]. Utilizing the knowledge that macromolecules have an af-
`finity for the lymphatic system, salts of four antibiotics were prepared with high-mo—
`lecular—weight polyacrylic acids, sulfonic or phosphorylated polysaccharides, and poly-
`uronic derivatives. Parenteral administration of these macromolecular salts produced low
`antibiotic blood levels for long periods while lymph levels were high. Since lymphatic
`circulation is slow, the preferential distribution of the antibiotics to the lymphatic sys-
`tem prolonged the passage through the body.
`The lauric acid salt of propranolol was studied as an alternative to polymeric for—
`' mula'tions for sustaining the release of propranolol HCl. The findings indicated that the
`laurate salt increased the bioavailability. This was attributed to micellar solubilization
`or ion-pairing which could lead to lymphatic absorption or lower efficiency of extrac-
`tion by the liver [20].
`Toxicity is reduced by choosing the appropriate salt form; two different strategies
`have been utilitzed to accomplish this. One is based on organic radicals that occur natu—
`rally and are readily excreted or metabolized. Using this approach, salts formed with
`choline [21—23], amino acids [24,25], and vitamins [24, 26—32] have been prepared that
`exhibit lower toxicity and fewer side effects than the parent molecule or other salts. The
`second strategy is to select a salt component that pharmacologically overcomes an un-
`favorable property or properties of the principal agent. Salts incorporating N—cyclo-
`hexylsulfamic acid, better known as cyclamates, can make bitter—tasting drugs accept-
`able because of their characteristic sweet taste. Cyclamate salts of dextromethorphan
`and chlorpheniramine [33] raise the bitterness thresholds compared to commonly oc—
`curring salts. The preparation and characterization of other cyclamic acid salts have been
`reported [34—37].
`Other examples include the preparation of the benzhydralamine salt of penicillin [38]
`and the 8—substituted theophylline salts of several antihistamines [39—42]. Benzhy—
`dralamine is an antihistamine. The preparation of the benzhydralamine salt of penicil—
`lin was an attempt to produce a repository form of penicillin with antiallergic proper-
`ties. The synthesis of the xanthine salts of several antihistamines was an attempt to
`counteract the drowsiness caused by the antihistamines with the stimulant properties of
`the xanthines. A number of other 8—substituted theophyllines have been prepared [21,
`43—49].
`
`Apotex Exhibit 1012.007
`
`Apotex Exhibit 1012.007
`
`

`

`458
`
`Salt Forms of Drugs and Absorption
`
`A quinidine salt with reduced toxicity has been prepared from polygalacturonic acid,
`a derivative of pectin [50,51]. This substance possesses special demulcent properties
`and inhibits mucosal irritation. It is used to reduce the shock to the gastrointestinal (GI)
`mucosa resulting from the liberation of irritating ions caused by the rapid dissociation
`of the conventional inorganic quinidine salts. Quinidine polygalacturonate is one—fourth
`as toxic orally as the sulfate.
`The N—(2-hydroxyethyl)pyrrolidine salt of diclofenac (DHEP) was prepared as part
`of a study to obtain salts with balanced hydrophilic and hydrophobic properties [52].
`Of the 24 salts synthesized, DHEP had the greatest solubility in both water and octanol.
`In addition, it exhibited surfactant properties and the ability to solubilize lipid materi—
`als above its critical micelle concentrations. These properties suggest that this salt is
`preferable to topical administration since it could promote its own absorption by inter-
`acting with the membrane components. Other compounds reported to be potentially
`useful as pharmaceutical salt forms are shown in Table 3.
`
`Physicochemical Studies
`
`Although different salts of the same drug elicit similar biological responses, the inten—
`sities of response may differ markedly [96,97]. A knowledge of the physicochernical
`properties of a salt and its influence on pharmacokineticsis necessary to understand the
`onset, duration, and intensity of action, relative toxicity, and possible routes of admin—
`istration [2]. The influence of salt form on volatility and hygroscopicity has been in—
`vestigated in preformulation studies [98].
`
`Solubility
`
`Solubility is an important factor in chemical stability, the formation of dosage forms,
`and the overall drug-absorption process.
`
`Common—Ion Efiect
`
`Hydrochloride salts are the most common anionic salt—forming species [7]. However,
`they do not necessarily enhance the solubility of poorly soluble basic drugs in a chlo-
`ride—containing medium because of the common—ion effect which suppresses the solu-
`bility product equilibrium [99—105]. In some instances, the solubility of various hydro—
`chlorides was less than that of the corresponding free base at gastric pH. The practical
`effect of reducing solubility could ultimately be a reduction of the dissolution rate in
`gastric juice. The Setschenow salting—out constants for chloride are highest for these
`slightly soluble hydrochlorides [106]. However, the relationship between the aqueous
`solubility of sparingly soluble salts and the empirical Setschenow saltingout constant
`is valid only at low concentrations of added salt [107].
`Prazosin1s an example of a drug with a strong chloride'1on dependence The hy—
`drochloride saltin water has a solubility of 1 4 mg/mL at 30° C, whereas1n 0 1M HCl
`it is 0.037 mg/mL [108].
`A common—ion effect on the sodium salt of an organic acid has also been reported
`[109]. The solubility and dissolution rates decreased with varying sodium ion concene
`
`Apotex Exhibit 1012.008
`
`Apotex Exhibit 1012.008
`
`

`

`Salt Forms of Drugs and Absorption
`
`459
`
`TABLE 3 Potentially Useful Salt Forms of Pharmaceutical Agents
`
`
`Salt—Forming Agent
`
`Compound Modified
`
`Property Modified
`
`Reference
`
`p—Acetamidobenzoic acid
`Acetylaminoacetic acid
`N—Acetyl—L—asparagine
`N—Acetylcystine
`
`Adamantoic acid
`
`Adipic acid
`
`N—Alkylsulfamates
`
`Anthraquinone—l ,5-
`disulfonic acid
`
`Arabogalactan sulfate
`(arabino)
`Arginine
`
`Aspartate
`Betaine
`
`Bis(2—carboxychr0mon—5-
`yloxy)a1kanes
`Carnitine
`4~Chloro—m—toluenesulfonic
`acid
`Decanoate
`
`Diacetyl sulfate
`Dibenzylethylenediamine
`Diethylamine
`Diguiacyl phosphate
`Dioctyl succinate
`Embonic (pamoic) acid
`
`Fructose—1,6-diphosphoric
`acid
`
`Glucose—l—phosphoric acid,
`Glucose—6—phosphoric acid
`L—Glutamine
`
`Hydroxynaphthoate
`2—(4—Imidazolyl)ethylamine
`Isobutanolamine
`Lauryl sulfate
`Lysine
`
`Mcthanesulfonic acid
`
`N—Methylglucamine
`
`Various amines
`
`Doxycycline
`Erythromycin
`Doxycycline
`
`Alkylbiguanides
`Piperazine
`
`Ampicillin
`Lincomycin
`Cephalexin
`
`Hygroscopicity
`Solubility
`Solubility, activity, stability
`Combined effect useful in
`
`pneumonia
`Prolonged action
`Stability, toxicity, organoleptic
`properties
`Absorption (oral)
`Solubility
`Stability, absorption
`
`53
`54
`55
`56
`
`57
`58
`
`59
`60
`61
`
`Various alkaloids
`
`Prolonged action
`
`62,63
`
`Cephalosporin
`Sulfobenzylpenicillin
`
`Erthromycin
`Tetracycline
`7—Aminoalkyltheophy—
`Hines
`Metformin
`
`Propoxyphene
`
`Heptaminol
`Thiamine
`
`Ampicillin
`Cephalosporins
`Tetracycline
`Vincamine
`
`Kanamycin
`Z—Phenyl—S—methyl-
`morpholine
`Tetracycline
`
`Erthromycin
`Tetracycline
`Erythromycin
`Erythromycin
`Bephenium
`Prostaglandin
`Theophylline
`Vincamine
`
`Sulfobenzylpenicillin
`
`Cephalosporin
`Pralidoxirne (2—PAM)
`Sulfobenzylpenicillin
`
`Toxicity
`Stability, hygroscopicity,
`toxicity
`Solubility
`Gastric absorption
`Activity, prolonged prophylactic
`effect
`
`Toxicity
`Organoleptic properties
`
`Proplonged action
`Stability, hygroscopicity
`Prolonged action
`Reduced pain on injection
`Activity
`Organoleptic properties
`Toxicity
`Toxicity
`
`Solubility
`
`Solubility
`Solubility
`Solubility
`Solubility, activity, stability
`Toxicity
`Prolonged action
`Stability
`Organoleptic properties
`Toxicity, stability, hygroscop—
`icity
`
`Solubility
`Toxicity, stability, hygroscop—
`icity
`
`64
`65
`
`66
`67
`68
`
`69
`70
`
`71
`72
`
`73,74
`75
`76
`77
`78
`79
`
`80
`
`80
`80
`80
`55
`81
`82
`83
`84
`65
`
`64
`85
`65
`
`Apotex Exhibit 1012.009
`
`Apotex Exhibit 1012.009
`
`

`

`460
`
`Salt Forms of Drugs and Absorption
`
`TABLE 3 (Continued)
`
`
`
`
` Salt—Forming Agent Compound Modified Property Modified Reference
`
`
`
`
`
`N—Methylpiperazine
`Morpholine
`2—Naphtha1enesulf0nic acid
`Octanoate
`Probenicid
`Tannic acid
`Theobromine acetic acid
`3,4,5—Trimeth0xybenzoate
`
`Tromethamine
`
`Reduced pain on injection
`Toxicity, faster onset of action
`Reduced pain on injection
`Organoleptic properties
`Prolonged action
`Organoleptic properties
`Prolonged action
`Activity
`Organoleptic properties
`Prolonged action
`Absorption (oral)
`Physical state
`
`75
`86
`75
`87
`71
`88
`89,90
`91
`92
`71
`93
`94
`
`Cephalosporins
`Phenylbutazone
`Cephalosporins
`Propoxyphene
`Heptaminol
`Pivampicillin
`Various amines
`Propoxyphene
`Tetracycline
`Heptaminol
`Aspirin
`Dinoprost
`(prostaglandin F)
`
`Salmeterol Local toleranceXinafoate 95
`
`
`
`
`
`trations. The reduction in solubility product in the presence of NaCl was attributed to
`a decrease in the degree of self—association of the drug in aqueous media.
`
`Formulation
`
`The choice of salt can have significant benefits for the formulation of a drug as, for
`example, with the cytotoxic drug, coralyne sulfoacetate. The solubility of coralyne
`chloride in water is 4.5 mg/mL, and that of the sulfoacetate is 6.5 mg/mL; however,
`solutions containing 25 mg/mL were required for iv infusion [110,111]. The solubility
`of the chloride salt was no higher in weakly alkaline aqueous media than in distilled
`water since it is a salt of a quaternary ammonium ion and the conjugate base of a strong
`acid. Adding sodium hydroxide greatly enhanced the solubility of the sulfoacetate. The
`reason is that the sulfoacetate anion is an acid which is ionized by the added base,
`resulting in an increase in the concentration of coralynium ion in solution.
`The solubility of a salt can influence the use of formulation adjuvants. In the pres—
`ence of methanesulfonic, acetic, and hydrochloric acids, 2,3,4,5—tetrahydro—8-
`(methylsulfonyl)—1-H~3~benzazepin—7—ol had water solubilities of approximately 440,. 320,
`and 1 mg/mL. Addition of sodium chloride to a saturated solution of the mesylate
`(methanesulfonic) salt, reduced the solubility to approximately 60 mg/mL, even with
`a sodium chloride concentration as low as 0.05 M. This was probably due to the rapid
`conversion of the mesylate to the hydrochloride salt and may preclude the use of so—
`dium chloride as an isoosmotic agent or the use of saline as diluent [103].
`In addition to its effect on solubility, the choice of salt is important to the useful-
`‘ ness and efficacy of the formulation. For example, hydrochloride salts in aqueous so—
`lution may lower'the pH, which can adversely affect their use in parenteral dosage forms
`because of the incidence of pain and subsequent venous inflammation [112]. It could
`also lead to incompatibilities with metal aerosol containers [108].
`
`Apotex Exhibit 1012.010
`
`Apotex Exhibit 1012.010
`
`

`

`Salt Forms of Drugs and Absorption
`
`461
`
`Sciarra et al.[113], using epinephrine as a model compound in an aerosol delivery
`system, points out that the solubility of the salt form in various propellants is impor-
`tant in products intended for local action in the lungs or for systemic therapy; further—
`more, the salt form should be soluble in extracellular fluids.
`
`Complex Salt Formation
`
`Organic acid salt forms of basic drugs, such as amines, frequently have higher aque—
`ous solubilities than their corresponding inorganic salts. Hydrochloric, nitric, sulfuric,
`and phosphoric salts of triamterine form insoluble complex salts [114]. Acetic acid pro—
`duced solubilities higher than those observed with any of the inorganic acids. Although
`acetic acid complexes with triamterine, an insoluble complex was not found. This is im—
`portant in the synthesis and selection of a salt form that exhibits enhanced bioavailability
`and desirable formulation characteristics.
`
`Studies have been conducted on the complexation of some drugs with sodium poly—
`phosphate [115]. Insoluble complex salts formed with amethocaine, amitriptyline, pro-
`pranolol, and verapamil, but not with atropine, ephedrine, and procaine. The complex
`salt formed with verapamil produced a prolonged dissolution profile in acid compared
`to pure verapamil, but because of hygroscopicity it was difficult to process and store.
`The solubility also of organic carboxylic acids is also affected by salt formation,
`in
`some cases adversely. For example, N—[4—(1,4-benzodioxan—6-yl)~2~thiazolyl] oxamic
`acid was less soluble in the presence of sodium, potassium, and calcium ions. How-
`ever, these ions increased the distribution coefficients significantly between water and
`l—octanol, even at low concentrations. The lower solubility was attributed to the for-
`mation of less soluble salts, whereas the increase in distribution coefficients was ex—
`plained by ion-pairing and/0r complexation [116].
`
`Solubility Predictions
`
`The solubility of a salt can be influenced by the structure of the organic moiety or by
`the hydrophilic properties of the anion or cation. A higher crystal lattice energy (crys—
`tallinity) is generally reflected by a higher melting point. An increase in melting point,
`usually by maximizing or encouraging crystal symmetry, reduces solubility. Gould [108]
`reports that the solubility of a drug frequently decreases by an order of magnitude with
`an increase of 100°C in its melting point. Where solubility and resultant pH are major
`issues, a low melting salt of a drug produced from a soluble, fairly weak acid or base,
`probably made in situ, is usually preferred.
`The increase or decrease in melting point of a series of salts of basic compounds
`depends on the controlling effect of crystallinity from the conjugate anion. This is
`exemplified by an experimental drug candidate, UK47880, which has a basic pKa of 8
`[108]. Salts prepared from planar, high melting aromatic sulfonic or hydroxycarboxylic
`acids yield high melting crystalline salts. However, flexible aliphatic acids such as cit-
`ric and dodecylbenzene sulfonic yielded oils. Gould [108] discussed how crystal lattice
`forces of drugs with good hydrogen bonding potential could be built up by consider—
`ing the symmetry and hydrogen bonding potential of the conjugate acid. He used epi~
`nephrine as an example, which gives high melting salts with small, strongly hydrogen-
`bonding acids like malonic and maleic. The larger bitartrate and presumably
`symmetrically unfavored fumarate give lower melting salts.
`
`Apotex Exhibit 1012.011
`
`Apotex Exhibit 1012.011
`
`

`

`462
`
`Salt Forms of Drugs and Absorption
`
`Various salts of oc-(2—piperidyl)—3,6-bis(trifluoromethyl)—9-phenanthrenemethanol
`[102], chlorhexidine [117], erythromycin [118], and the N—alkylsulfamates of lincomycin
`[60] show enhanced solubility which can be attributed to a lower melting point and the
`hydrophilic properties of the anion. Organic salts may increase aqueous solubility
`through decreased crystal lattice energy, lowered melting point, increased hydrogen
`bonding of the salt counterions with water, etc.
`There are exceptions to the solubility—melting point and solubility—hydrophilicity
`relationship. For example, the THAM, tris(hydroxymethyl)aminomethane, salts of
`certain analgesic—anti-inflammat0ry agents showed no simple solubility—melting point
`relationship [119]. Anderson and Condradi, using organic amine salts of flurbiprofen
`to predict water solubility, found a strong dependence of the solubility product on melt—
`ing point; however, there was no significant correlation between solubility product and
`counterion hydrophilicity [120]. The authors concluded that this is in conflict with the
`notion that higher salt solubilities can be achieved by selecting more hydrophilic
`‘ counterions, since such arguments neglect the likelihood that interactions in the crystal
`become stronger as the salt-forming species are made increasingly polar.
`Rubino [121] found that the logarithms of the molar solubilities of a number of
`sodium salts of drugs were inversely related to their melting points, but a good. corre-
`lation was not evident. However, the logarithms of the molar solubilities were inversely
`related to both the melting points and stoichiometric amounts of water in the crystal
`hydrates, but unrelated to the polarity of the corresponding acid forms of the drugs. It
`was concluded therefore that the melting point and the degree of crystal hydration of
`the solid phase are most important in determining the solubilities of the sodium salts
`of some drugs.
`The solubilities of the sodium salts of some weakly acid drugs have been determined
`in mixtures of propylene glycol and water. The solubility in the mixed solvent of com—
`pounds with low temperatures of desolvation had increased, whereas the solubility of
`compounds with high desolvation temperatures had decreased. These data indicate that
`crystal hydrate formation plays a significantrole in determining if a cosolvent can be
`used to enhance the solubilities of certain sodium salts [122].
`The hydrogen ion concentration can significantly affect salt solubility. Anderson
`[123] discussed the influence of pH on the solubility of therapeutically useful weak acids
`and bases and their salts. This was followed a few years later by an extensive study on
`the solubility interrelationships of the hydrochloride and free base of two amines [124].
`Mathematical equations describing the total solubility at an arbitrary pH in terms of the
`independent solubilities of the hydrochloride and free base species and the dissociation
`constant of the salt were derived and fitted to experimental data with good results. This
`report made the point that, although the solubility of an amine hydrochloride generally
`sets the maximum obtainable concentration for a given amine, the solubility of the free
`base and the pKa determine the maximum pH at which formulation as a solution is pos—
`sible. This assumes that the desired concentration exceeds the free base solubility.
`Shifting the pH—solubility profile to higher pH values for formulation purposes may
`require increasing the solubility of the free base with the help of an appropriate co—
`solvent. Because the dissociation characteristics of carboxylic acids and other organic
`species are similar to those of organic hydrochlorides, the pH—solubility profiles could
`be characterized theoretically by the same treatment.
`Chowhan [125] studied the solubilities of three organic carboxylic acids (naproxen;
`7-methylsulfinyl-2~xanthonecarboxylic acid; and 7—methythio—2-xanthonecarboxylic acid)
`and their sodium, potassium, calcium, and magnesium salts as a function of pH. The
`
`Apotex Exhibit 1012.012
`
`Apotex Exhibit 1012.012
`
`

`

`Salt Forms of Drugs and Absorption
`
`463
`
`data were fitted to mathematical relationships similar to those used by Kramer and Flynn
`[124]. The results on the solubility of naproxen and its salts were in excellent agree—
`ment with theory. The solubilities of the two xanthone carboxylic acids and their salts
`were higher at higher pH than the values calculated for complete dissociation in solu—
`tion.
`
`Surface Activity
`
`The salts of some compounds are surface active [126—128]. If the saturation solubility
`enables the critical micelle concentration (CMC) to be reached, solubility is enhanced
`significantly via micellar solubilization. A study of the colloidal properties of some
`chlorhexidine salts showed that the counterion can affect the CMC which was usually
`associated with a change in micellar size [126]. For example, the diacetate displays a
`higher CMC than the digluconate [126].
`The hydrochloride salt of 2—butyl-3—benzofuranyl—4—[2-(diethylamino)eth0xy]~3,5-
`diiodophenyl ketone is capable of forming micelles. Anions such as chloride, sulfate,
`acetate, tartrate, and citrate significantly affect the equilibrium solubility of the com—
`pound, which is partly related to the effect on the CMC by the anionic environment
`[127].
`The nonopioid kappa agonist analgesic amine, DuP 747, as the hydrochloride salt,
`exerts surface activity in aqueous solutions; however, the critical micellar concentra—
`tion is not reached at the saturation solubility [128]. On the other hand, the methane-
`sulfonate salt formed a micellar solution and allowed for a solubility of 60 mg/mL as
`opposed to 3 mg/mL for the hydrochloride.
`Zomepirac, an insoluble, carboxylic, non-narcotic analgesic, has a solubility in water
`of 0.02 mg/mL. In a developing zomepirac solution containing 100 mg/mL [129],
`THAM was found to be a satisfactory solubilizer at a concentration where equivalent
`concentrations of sodium or potassium hydroxide were not. The solubility was achieved
`by a micellar mechanism. It is interesting that potassium hydroxide was more effective
`in solubilizing zomepirac than sodium hydroxide. Walkling et al. attributed the difference
`in their performance as solubilizers to the difference in their charge densities [129]. Ad-
`ditional references on the relationship of salt form and solubility are listed in Table 4.
`
`Dissolution Rate
`
`In many cases, the dissolution rate can be a good indicator of bioavailability, especially
`of poorly soluble drugs. A salt form frequently exhibits a higher dissolution rate than
`the corresponding conjugate acid or base at the same pH, even though they may have
`the same equilibrium solubility. In a review article on the biopharmaceutical basis for
`drug design, Nelson [150,151], and later Benet [152], referred to the self—buffering
`action of the salt form in the diffusion layer. The dissolution rates are determined by
`the pH values of the diffusion layer and are independent of the prulk of the media used.
`Therefore, the difference in diffusion—layer pH between a parent compound and its salt
`accounts for the difference in the dissolution rates in a particular medium.
`
`Effect of Salt Form
`
`Nelson, using theophylline salts, was the first to show the correlation between diffu—
`sion—layer pH and dissolution rate [150]. Salts with a high diffusion-layer pH had higher
`
`Apotex Exhibit 1012.013
`
`Apotex Exhibit 1012.013
`
`

`

`464
`
`Salt Forms of Drugs and Absorption
`
`TABLE 4 References on Salt Form and Solubility
`
`
`
` Topic Reference
`
`Mineral acid salts of lidocaine
`Nonionic surfactant effect on rate of release of drugs from suppositories
`Influence of solubility of salicyclic acid on diffusion from ointment bases
`Influence of solubility on rate of GI absorption of aspirin
`Effect of dosage form on G1

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