throbber
Journal of
`Pharmaceutical
`Sciences
`
`JANUARY 1977
`VOLUME 66 NUMBER 1
`
`REVIEW ARTICLE
`
`Pharmaceutical Salts
`
`STEPHEN M. BERGE **, LYLE D. BIGHLEY *, and
`DONALD C. MONKHOUSE
`
`Keyphrases 0 Pharmaceutical salts-general
`pharmacy, physico-
`chemical properties, bioavailability, pharmaceutical properties, toxi-
`Salts, pharmaceutical-general
`pharmacy, physico-
`cology, review
`chemical properties, bioavailability, pharmaceutical properties, toxi-
`Physicochernical properties-dissolution,
`solubility,
`cology, review
`stability, and organoleptic properties of pharmaceutical salts, review
`Bioavailability-formulation
`effects, absorption alteration and phar-
`macokinetics of pharmaceutical salts, review 0 Toxicology-pharma-
`ceutical salts, review
`
`2
`4
`5
`7
`
`CONTENTS
`Potentially Useful Salts ..................................
`Physicochemical Studies ........................
`..........................
`Dissolution Rate ....
`Solubility ..........
`..........................
`Organoleptic Properties .........................
`...............................
`s Properties ....................
`.....................
`.....................
`Absorption Alteration . .
`.........................
`General Pharmacy .......................................
`14
`..........
`.......... 14
`Pharmacological Effect
`Dialysis ...............................................
`14
`Miscellaneous .........................................
`14
`Toxicological Considerations ..............................
`15
`Toxicity of Salt Ion .....................................
`15
`Toxicity of Salt Form ...................................
`15
`Conclusions .............................................
`16
`References ..............................................
`16
`
`10
`11
`11
`
`The chemical, biological, physical, and economic char-
`acteristics of medicinal agents can be manipulated and,
`hence, often optimized by conversion to a salt form.
`Choosing the appropriate salt, however, can be a very
`difficult task, since each salt imparts unique properties to
`the parent compound.
`
`Salt-forming agents are often chosen empirically. Of the
`many salts synthesized, the preferred form is selected by
`pharmaceutical chemists primarily on a practical basis:
`cost of raw materials, ease of crystallization, and percent
`yield. Other basic considerations include stability, hy-
`groscopicity, and flowability of the resulting bulk drug.
`Unfortunately, there is no reliable way of predicting the
`influence of a particular salt species on the behavior of the
`parent compound. Furthermore, even after many salts of
`the same basic agent have been prepared, no efficient
`screening techniques exist to facilitate selection of the salt
`most likely to exhibit the desired pharmacokinetic, solu-
`bility, and formulation profiles.
`Some decision-making models have, however, been de-
`veloped to help predict salt performance. For example,
`Walkling and Appino (1) described two techniques, “de-
`cision analysis” and “potential problem analysis,” and
`applied them to the selection of the most suitable deriva-
`tive of an organic acid for development as a tablet. The
`derivatives considered were the free acid and the potassi-
`um, sodium, and calcium salts. Both techniques are based
`on the chemical, physical, and biological properties of these
`specific derivatives and offer a promising avenue for de-
`veloping optimal salt forms.
`Information on salts is widely dispersed throughout the
`pharmaceutical literature, much of which addresses the
`use of salt formation to prolong the release of the active
`component, thereby eliminating various undesirable drug
`properties (24). This review surveys literature of the last
`25 years, emphasizing comparisons between the properties
`of different salt forms of the same compound. Included also
`is a discussion of potentially useful salt forms. Our purpose
`is twofold: to present an overview of the many different
`salts from which new drug candidates can be chosen and
`
`Vol. 66, No. 1 , January 19771 1
`
`Liquidia's Exhibit 1034
`IPR2020-00770
`Page 1
`
`

`

`Table I-FDA-Approved Commercially Marketed Salts
`Anion
`Percent”
`
`Acetate
`Benzenesulfonate
`Benzoate
`Bicarbonate
`Bitartrate
`Bromide
`Calcium edetate
`CamsyIateh
`Carbonate
`Chloride
`Citrate
`Dihydrochloride
`Edetate
`EdisylateC
`Estolate
`Esylate“
`Fumarate
`Gluceptatef
`Gluconate
`Glutamate
`Glycollylarsanilateg
`Hexylresorcina te
`Hydrabamine”
`Hydrobromide
`Hydrochloride
`H ydroxynaphthoate
`
`Cation
`
`1.26
`0.25
`0.51
`0.13
`0.63
`4.68
`0.25
`0.25
`0.38
`4.17
`3.03
`0.51
`0.25
`0.38
`0.13
`0.13
`0.25
`0.18
`0.51
`0.25
`0.13
`0.13
`0.25
`1.90
`42.98
`0.25
`
`Anion
`
`lodide
`Isethionate‘
`Lactate
`Lactobionate
`Malate
`Maleate
`Mandelate
`Mesylate
`Methylbromide
`Methylnitrate
`Methylsulfate
`Mucate
`Napsylate
`Nitrate
`Pamoate (Embonate)
`Pantothenate
`Phosphateldiphosphate
`Polygalacturonate
`Salicylate
`Stearate
`Subacetate
`Succinate
`Sulfate
`Tannate
`Tartrate
`Teoclate’
`Triethiodide
`
`Percenta
`
`2.02
`0.88
`0.76
`0.13
`0.13
`3.03
`0.38
`2.02
`0.76
`0.38
`0.88
`0.13
`0.25
`0.64
`1.01
`0.25
`3.16
`0.13
`0.88
`0.25
`0.38
`0.38
`7.46
`0.88
`3.54
`0.13
`0.13
`
`Percent“
`
`Cation
`
`Percent“
`
`Organic:
`B e n z a t h i d
`Chloroprocaine
`Choline
`Diethanolamine
`Ethylenediamine
`Meglumine‘
`Procaine
`Camphorsulfonate. 1,2-Ethanedisulfonate.
`Lauryl sulfate.
`0 Percent is based on total number of anionic or cationic salts in use through 1974.
`c Ethanesulfonate. f Glucoheptonate. g p-Glycollamidophenylarsonate. N,N’-Di(dehydroabiety1)ethylenediamine. 2-Hydroxyethanesulfonate.
`1 8-C hlorotheophyllinate. N,N‘-Dibenzylethylenediamine. N- Met,hylglucamine.
`
`0.66
`0.33
`0.33
`0.98
`0.66
`2.29
`0.66
`
`Metallic:
`Aluminum
`Calcium
`Lithium
`Magnesium
`Potassium
`Sodium
`Zinc
`
`0.66
`10.49
`1.64
`1.31
`10.82
`61.97
`2.95
`
`to assemble data that will provide, for the student and
`practitioner alike, a rational basis for selecting a suitable
`salt form.
`
`POTENTIALLY USEFUL SALTS
`Salt formation is an acid-base reaction involving either
`a proton-transfer or neutralization reaction and is there-
`fore controlled by factors influencing such reactions.
`Theoretically, every compound that exhibits acid or base
`characteristics can participate in salt formation. Particu-
`larly important is the relative strength of the acid or
`base-the acidity and basicity constants of the chemical
`species involved. These factors determine whether or not
`formation occurs and are a measure of the stability of the
`resulting salt.
`The number of salt forms available to a chemist is large;
`surveys of patent literature show numerous new salts being
`synthesized annually. Various salts of the same compound
`often behave quite differently because of the physical,
`chemical, and thermodynamic properties they impart to
`the parent compound. For example, a salt’s hydrophobicity
`and high crystal lattice energy can affect dissolution rate
`and, hence, bioavailability. Ideally, it would be desirable
`if one could predict how a pharmaceutical agent’s prop-
`erties would be affected by salt formation.
`Tables I and I1 list all salts that were commercially
`marketed through 1974. The list was compiled from all
`agents listed in “Martindale The Extra Pharmacopoeia,”
`
`2 1 Journal of Pharmaceutical Sciences
`
`26th ed. (7). Table I categorizes all salt forms approved by
`the Food and Drug Administration (FDA), while Table I1
`lists those not approved by the FDA but in use in other
`countries. (Only salts of organic compounds are considered
`because most drugs are organic substances.) The relative
`frequency with which each salt type has been used is cal-
`culated as a percentage, based on the total number of an-
`ionic or cationic salts in use through 1974. Because of
`simple availability and physiological reasons, the mono-
`protic hydrochlorides have been by far the most frequent
`choice of the available anionic salt-forming radicals, out-
`numbering the sulfates nearly six to one. For similar rea-
`sons, sodium has been the most predominant cation.
`Knowledge that one salt form imparts greater water
`solubility, is less toxic, or slows dissolution rate would
`greatly benefit chemists and formulators. In some cases,
`such generalizations can be made. Miller and Heller (8)
`discussed some properties associated with specific classes
`of salt forms. They stated that, in general, salt combina-
`tions with monocarboxylic acids are insoluble in water and
`lend themselves to repository preparations, while those of
`dicarboxylic acids confer water solubility if one carboxylic
`group is left free. Pamoic acid, an aromatic dicarboxylic
`acid, is an exception since it is used as a means of obtaining
`prolonged action by forming slightly soluble salts with
`certain basic drugs. Saias et al. (9) reviewed the use of this
`salt form in preparing sustained-release preparations.
`More recently, latentiation of dihydrostreptomycin (10)
`
`Liquidia's Exhibit 1034
`IPR2020-00770
`Page 2
`
`

`

`Table II-Non-FDA- Approved Commercially Marketed
`Salts
`
`Anion
`
`Adipate
`Alginate
`Aminosalicylate
`An hydromethylenecitrate
`Arecoline
`Aspartate
`Bisulfate
`Butylbromide
`Camphorate
`Digluconate
`Dihydrobromide
`Disuccinate
`Glycerophosphate
`Hemisulfate
`Hydrofluoride
`Hydroiodide
`Methylenebis(salicy1ate)
`Napadisylate
`Oxalate
`Pectinate
`Persulfate
`Phenylethylbarbiturate
`Picrate
`Propionate
`Thiocyanate
`Tosylate
`Undecanoate
`
`PPrrPnta
`
`0.13
`0.13
`0.25
`0.13
`o.i3
`0.25
`0.25
`0.13
`0.13
`0.13
`0.13
`0.13
`0.88 o.i3
`0.13
`0.25
`0.13
`0.13
`0.26
`_.
`0.13
`0.13
`0.13
`0.13
`0.13
`0.13
`0.13
`0.13
`
`~
`
`Percent a
`
`Cation
`Organic:
`Benethamine
`Ckmizoled
`Diethylamine
`Piperazine
`Tromethamine‘
`Metallic:
`Barium
`0.33
`0.98
`Bismuth
`a Percent is based on total number of anionic and cationic salts in use
`through 1974. 1,5-Naphthalenedisulfonate. N-Benzylphenethylamine.
`l-p-Chlorobenzyl-2-pyrrolidin-l’-ylmethylbenzimidazole. Tris(hy-
`droxymethy1)aminomethane.
`
`0.33
`0.33
`0.33
`0.98
`0.33
`
`using pamoic acid resulted in the formation of a delayed-
`action preparation. Numerous studies using pamoate salts
`are dispersed throughout the literature (11-15).
`Alginic acid also has been used to prepare long-acting
`pharmaceuticals. Streptomycin alginate was prepared (16)
`and shown to be effective in sustained-release prepara-
`tions. A striking example of a long-acting alginate salt is
`that of pilocarpine. When dispersed in sterile water and
`dried to a solid gel, this compound was found useful in the
`preparation of long-acting ophthalmic dosage forms (17).
`While liquid preparations of the alginate and hydrochlo-
`ride salts possess similar miotic activity, studies showed
`that solid pilocarpine alginate flakes constricted pupil size
`more effectively and increased the duration of miosis sig-
`nificantly when compared with the liquid preparations.
`Solid dose pilocarpine may be more uniformly available,
`because it diffuses more slowly through the gel matrix
`which holds the drug in reserve. In contrast, drops of the
`commonly employed solution dosage form release the dose
`immediately to the conjunctival fluid.
`MQlek et al. (18) devised a unique way of prolonging
`action through salt formation; they showed that the dis-
`tribution of several antibiotics could be markedly altered
`by merely preparing macromolecular salts. Since macro-
`molecules and colloidal particles have an affinity for the
`lymphatic system, streptomycin, neomycin, viomycin, and
`
`streptothrycin were combined with high molecular weight
`compounds such as polyacrylic acids, sulfonic or phos-
`phorylated polysaccharides, and polyuronic derivatives.
`Parenteral administration of these compounds produced
`low blood levels of the antibiotic for long periods, while
`lymph levels were high. (In comparison, streptomycin
`sulfate gave high blood levels but low lymph levels.) This
`alteration in distribution caused the streptomycin to
`prolong its passage through the body, since lymphatic
`circulation is quite slow.
`The appropriate choice of a salt form has been found to
`reduce toxicity. It can be rationalized that any compound
`associated with the normal metabolism of food and drink
`must be essentially nontoxic. The approach of choosing
`organic radicals that are readily excreted or metabolized
`opened up a new class of substances from which to select
`a salt form. For example, certain salts of the strong base
`choline have proven to be considerably less toxic than their
`parent compound. The preparation and properties of
`choline salts of a series of theophylline derivatives were
`reported (19), and it was shown that choline theophyllinate
`possessed a greater LD50 than theophylline or its other
`salts (20). It was postulated that this agent would be less
`irritating to the GI tract than aminophylline, because “its
`basic constituent, choline, is an almost completely non-
`toxic substance of actual importance to the physiologic
`economy.” This evidence led to the preparation of choline
`salicylate (21) as an attempt to reduce the GI disturbances
`associated with salicylate administration. Clinical studies
`indicated that choline salicylate elicited a lower incidence
`of GI distress, was tolerated in higher doses, and was of
`greater benefit to the patient than was acetylsalicylic acid
`(aspirin).
`Amino acids and acid vitamins also have been used as
`salt-forming agents. Based on the evidence that coad-
`ministration of amino acids with aminoglycoside antibi-
`otics reduced their toxicity, a series of amino acid salts of
`dihydrostreptomycin was prepared (22). In all but one
`case, the acute toxicities of these salts were lower than the
`toxicity of the sulfate. The ascorbate and pantothenate
`also were synthesized and shown to be less toxic than the
`sulfate. Of the salts prepared, the ascorbate had the highest
`LD50.
`The vitamins most commonly used for forming salts
`exhibiting reduced toxicity are ascorbic and pantothenic
`acids. Keller et al. (23) were the first to use pantothenic
`acid as a means of “detoxifying” the basic streptomyces
`antibiotics. Parenteral administration of the pantothen-
`ates of streptomycin and dihydrostreptomycin had a sig-
`nificantly reduced incidence of acute neurotoxicity in cats
`as compared with the sulfates. Subsequent studies (24-28)
`supported this finding and showed that the pantothenates
`of neomycin and viomycin also are less toxic. The ascorbate
`of oleandomycin was synthesized and its pharmacological
`properties were reported (29). Upon intramuscular injec-
`tion in rats, it produced less irritation than the phos-
`phate.
`p -Acetamidobenzoic acid, an innocuous metabolite of
`folk acid present in normal blood and urine, has been used
`in preparing salts. In particular, it yields stable salts with
`amines that otherwise tend to form hygroscopic products
`with conventional acid components (30).
`Often the salt form is chosen by determining a salt
`
`Val. 66, No. 1, January 1977 1 3
`
`Liquidia's Exhibit 1034
`IPR2020-00770
`Page 3
`
`

`

`component that will pharmacologically antagonize an
`unfavorable property or properties exhibited by the basic
`agent. Salts of N-cyclohexylsulfamic acid are an example
`of the practical application of this approach. N-Cyclo-
`hexylsulfamic acid salts, better known as cyclamates, have
`a characteristic sweet, pleasing taste. Although presently
`under investigation by the FDA for potentially carcino-
`genic properties, salts incorporating this compoynd can
`render unpleasant or bitter-tasting drugs acceptable. For
`example, the cyclamates of dextromethorphan and
`chlorpheniramine exhibit greatly improved bitterness
`thresholds compared to commonly occurring salts (31).
`Furthermore, their stability in aqueous solution was de-
`scribed as good when mtiintained at a pH not greater than
`4.
`N-Cyclohexylsulfamic acid salts of thiamine hydro-
`chloride and lincomycin also have been synthesized. Thi-
`amine N-cyclohexylsulfamate hydrochloride was reported
`to have a more pleasant taste than other thiamine salts
`while having an equal or greater stability (32). Lincomycin
`cyclamate, shown to possess an enhanced thermal stability
`over its hydrochloride, was prepared (33) to test the hy-
`pothesis that reduced lincomycin absorption in the pres-
`ence of small quantities of cyclamates was due to a simple
`metathetic reaction. However, this assumption was found
`not to be true. An extensive study of the preparation and
`characterization of cyclamic acid salts of several widely
`used classes of drugs including antihistamines, antibiotics,
`antitussives, myospasmolytics, and local anesthetics was
`reported (34,35).
`Various salts of penicillin and basic amine compounds
`have been formulated in an effort to produce a long-acting,
`nonallergenic form of penicillin. Since antihistamines
`appear to mitigate the symptomatology of penicillin re-
`actions in some patients, coadministration of the two has
`been advocated. The preparation of the benzhydralamine
`salt of penicillin was an attempt to produce a repository
`form of penicillin with antiallergic properties (36). Blood
`levels achieved with this salt were comparable to those of
`penicillin G potassium; however, its antiallergic properties
`were not evaluated. In fact, the investigators noted that
`antihistamines can actually cause sensitization at times
`and stated that “despite their occasionally favorable in-
`fluence on the symptoms of penicillin sensitivity, they
`contribute directly to the potential of drug sensitivity when
`co-administered with penicillin.’’
`Silver salts of sulfanilamide, penicillin, and other anti-
`biotics have been prepared and represent cases where the
`species (ions) are complementary. When aqueous solutions
`of the salts were applied topically to burned tissue, they
`yielded the combined benefits of the oligodynamic action
`of silver and the advantages of the antibacterial agents
`(37).
`The use of 8-substituted xanthines, particularly the
`8-substituted theophyllines, as salt-forming agents was
`first reported in the preparation of a series of antihistamine
`salts (38-41). Synthesis of these xanthine salts was an at-
`tempt to find a drug to counteract the drowsiness caused
`by the antihistamines with the stimulant properties of the
`xanthines. When an electronegative group is introduced
`into the xanthine molecule at the 8-position, the elec-
`tron-drawing capacity of the substituent results in the
`creation of an acidic hydrogen at position 7. Thus, these
`
`4 /Journal of Pharmaceutical Sciences
`
`moderately strong acidic compounds can undergo salt
`formation with various organic bases.
`The 8-halotheophyllines were the first group of xan-
`thines studied as potential salt-forming agents. Since the
`report on the preparation of the 8-chlorotheophylline salt
`of diphenhydramine (42), synthesis of the 8-halotheo-
`phyllinates of a number of organic bases has been at-
`tempted. The 8-chlorotheophylline salts of quinine,
`ephedrine, and strychnine were prepared and character-
`ized (43). These salts were less water soluble than the
`corresponding free alkaloidal bases. In a similar report, the
`8-chlorotheophyllinates of three synthetic narcotics,
`meperidine, levorphanol, and metopon, were prepared
`(44).
`Pharmacological and clinical studies involving the 8-
`bromotheophylline pyrilamine salt revealed the unusual
`diuretic properties associated with the 8-halotheophylline
`portion of the compound (45,46). This finding initiated
`an investigation into the preparation of a soluble 8-bro-
`motheophylline salt of high diuretic activity. With readily
`available amines, over 30 salts. were synthesized and
`screened for diuretic activity (47). When tested against
`theophylline salts of the same amines, the 8-bromotheo-
`phyllinates showed greater activity in every case.
`With the successful formation of 8-halotheophyllinates
`of organic bases, Morozowich and Bope (48) proposed that,
`if the halogen moiety was replaced with a more electro-
`negative substituent such as a nitro group, a more acidic
`compound would be formed. Presumably, more stable salts
`would result and precipitation of the free xanthine deriv-
`ative in the stomach would be less likely to occur. On this
`premise, they successfully prepared pharmacologically
`effective 8-nitrotheophyllinates of several pharmaceuti-
`cally useful bases.
`Duesel et al. (19), in their study of choline theophylli-
`nate, prepared the 8-chloro-, 8-bromo-, and 8-nitrotheo-
`phylline salts of choline. Oral toxicity studies in mice
`showed that the LD.50 of the 8-nitrotheophyllinate was
`much greater than that of either 8-halotheophylline. In
`fact, it remained nonlethal at doses as high as 5 g.
`Polygalacturonic acid, a derivative of pectin, has been
`used to prepare quinidine salts exhibiting reduced toxicity
`(49, 50). The compound possesses special demulcent
`properties and inhibits mucosal irritation. The rationale
`for use of this agent is to reduce the ionic shock to the GI
`mucosa resulting from the flood of irritating ions liberated
`by rapid dissociation of the conventional inorganic quin-
`idine salts. Studies have shown that it is four times less
`toxic orally than the sulfate. This difference was attributed
`to the slower release of quinidine from the polygalactu-
`ronate.
`Other compounds reported to be potentially useful as
`pharmaceutical salt forms are listed in Table 111.
`
`PHYSICOCHEMICAL STUDIES
`
`Biological activity of a drug molecule is influenced by
`two factors: its chemical structure and effect at a specific
`site and its ability to reach-and
`then be removed from-
`the site of action. Thus, a knowledge of the physicochem-
`ical properties of a compound that influence its absorption,
`distribution, metabolism, and excretion is essential for a
`complete understanding of the onset and duration of ac-
`
`Liquidia's Exhibit 1034
`IPR2020-00770
`Page 4
`
`

`

`Table 111-Potentially Useful Salt Forms of Pharmaceutical Agents
`
`Salt-Forming Agent
`
`Compound Modified
`
`Modification
`
`Reference
`
`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-carboxychromon-5-yloxy)alkanes
`.
`-
`
`Carnitine
`4-Chloro-rn-toluenesulfonic acid
`Decanoate
`Diacetyl sulfate
`Dibenzylethylenediamine
`Diethylamine
`Diguaiacyl phosphate
`Dioctyl sulfosuccinate
`Embonic (pamoic) acid
`Fructose 1,6-diphosphoric acid
`
`Glucose 1-phosphoric acid, glucose
`6-phosphpric acid
`I,-Glutamine
`H ydroxynaphthoate
`2-( 4-Imidazoly1)ethylamine
`Isobutanolamine
`Lauryl sulfate
`Lysine
`
`Methanesulfonic acid
`N - Methylglucamine
`N-Methylpiperazine
`Morpholine
`2-Naphthalenesulfonic acid
`Octanoate
`Probenecid
`Tannic acid
`Theobromine acetic acid
`3,4,5-Trimethoxybenzoate
`
`Tromethamine
`
`Doxycycline
`Erythromycin
`Doxycycline
`A1 kylbiguanides
`Piperazine
`Ampicillin
`Lincomycin
`Cephalexin
`Various alkaloids
`Cephalosporins
`a-Sulfobenzylpenicillin
`Erythromycin
`Tetracycline
`7-(Aminoalkyl) theophyllines
`Metformin
`Propoxyphene
`Heptaminol
`Thiamine
`Ampicillin
`Cephalosporins
`Tetracycline
`Vincamine
`Kanamycin
`2-Phenyl-3-methylmorpholine
`Tetracycline
`Erythromycin
`Tetracycline
`Erythromycin
`Erythromycin
`Bephenium
`Prostaglandin
`Theophylline
`Vincamine
`a-Sulfobenzylpenicillin
`Cephalosporins
`Pralidoxime (2-PAM)
`a-Sulfobenzylpenicillin
`Cephalosporins
`Phenylbutazone
`Cephalosporins
`Propoxyphene
`Heptaminol
`Pivampicillin
`Various amines
`Propoxyphene
`Tetracycline
`Heptaminol
`Aspirin
`DinoDrost (mostadandin F d
`
`Solubility
`Solubility, activity, stability
`Combined effect useful in pneumonia
`Prolonged action
`Stability, toxicity, organoleptic properties
`Absorption (oral)
`Solubility
`Stability, absorption
`Prolonged action
`Toxicity
`Stability, hygroscopicity, toxicity
`Solubility
`Gastric absorption
`Activity, prolonged prophylactic effect
`Toxicity
`Organoleptic properties
`Prolonged 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, hygroscopicity
`
`Solubility
`Toxicity, stability, hygroscopicity
`Reduced Dain on iniection
`Toxicity, ‘faster o n k t of action
`Reduced pain on injection
`Organoleptic properties
`Prolonged action
`Organoleptic properties
`Prolonged action
`Activity
`Organoleptic properties
`Prolonged action
`Absorption (oral)
`Phvsical state
`
`51
`52
`53
`54
`55
`56
`57
`58
`59,60
`61
`62
`63
`64
`65
`66
`67
`68
`69
`70,71
`72
`73
`14
`75
`76
`77
`
`77
`
`52
`78
`79
`80
`81
`62
`61
`82
`62
`72
`83
`72
`84
`68
`85
`86,87
`88
`89
`68
`90
`91
`
`tion, the relative toxicity, and the possible routes of ad-
`ministration (2).
`In a review in 1960, Miller and Holland (92) stated that
`“different salts of the same drug rarely differ pharmaco-
`logically; the differeaces are usually based on the physical
`properties.” In a subsequent review (931, Wagner ex-
`panded upon this statement, asserting that, although the
`nature of the biological responses elicited by a series of
`salts of the same parent compound may not differ appre-
`ciably, the intensities of response may differ markedly.
`The salt form is known to influence a number of physi-
`cochemical properties of the parent compound including
`dissolution rate, solubility, stability, and hygroscopicity.
`These properties, in turn, affect the availability and for-
`mulation characteristics of the drug. Consequently, the
`pharmaceutical industry has systematically engaged in
`extensive preformulation studies of the physicochemical
`properties of each new drug entity to determine the most
`suitable form for drug formulation. Published information
`concerning such studies, however, is sparse. Preformula-
`tion studies have been outlined, and the influence of the
`salt form on the volatility and hygroscopicity of an agent
`under investigation was discussed (94).
`
`In one such study, methylpyridinium-2-aldoxime
`(pralidoxime) salts were investigated (95). This study set
`out to prepare a salt with water solubility adequate to allow
`intramuscular injection of a low volume (2-3 ml) thera-
`peutic dose. The original compound, the methiodide, had
`the disadvantages of limited aqueous solubility and high
`potential toxicity, since its high iodide content could result
`in iodism. On the basis of physiological compatibility,
`better water solubility, favorable stability, and relatively
`high percentage of oxime, the chloride salt of pralidoxime
`was selected for therapeutic administration; it was claimed
`that “the anion used to form the salt can confer physical
`properties of importance and significance for the formu-
`lation and administration of the compound” (95).
`Some physicochemical properties of a series of mineral
`acid salts of lidocaine also were determined (96). While the
`hydrochloride and hydrobromide were more hygroscopic,
`they were more soluble in a number of solvents than the
`nitrate; perchlorate, phosphate, or sulfate salts.
`Dissolution Rate-The dissolution rate of a pharma-
`ceutical agent is of major importance to the formulator. In
`many cases, particularly with poorly soluble drugs, this
`characteristic best reflects the bioavailability of the com-
`
`Vol. 66, No. 1, January 1977 1 5
`
`Liquidia's Exhibit 1034
`IPR2020-00770
`Page 5
`
`

`

`pound. As a rule, a pharmaceutical salt exhibits a higher
`dissolution rate than the corresponding conjugate acid or
`base at an equal pH, even though they may have the same
`equilibrium solubility. The explanation for this result lies
`in the processes that control dissolution.
`Dissolution can be described by a diffusion layer modell
`in terms of an equation developed by Nernst and Brunner
`(97):
`
`..
`
`(Eq. 1)
`where W is the mass of the solute dissolved at time t,
`d W l d t is the rate of mass transfer per unit time, D is the
`solute molecule diffusion coefficient, S is the surface area
`of the dissolving solid, h is the diffusion layer thickness,
`C is the concentration of the drug in the bulk solution at
`time t , and C,s is the saturation solubility of the solute in
`the diffusion layer.
`The driving force for dissolution in Eq. 1 is the difference
`between the saturation solubility of the drug and the
`concentration of the drug in the bulk fluid. If the drug is
`not rapidly absorbed after it dissolves, then C, the con-
`centration in the bulk solution, approaches C, and the
`dissolution rate is retarded. When this occurs, absorption
`is “absorption rate” limited (or “membrane transport”
`limited). If the absorption rate is rapid (or if the absorption
`mass transfer coefficient is much larger than DSlh of Eq.
`1)’ however, C becomes negligible compared to C, and
`dissolution occurs under “sink” conditions. Absorption is
`then said to be dissolution rate limited, which is what oc-
`curs with most poorly soluble drugs. In either case, an in-
`crease in C,, as in salt formation, increases dissolution.
`Salts often speed dissolution by effectively acting as
`their own buffers to alter the pH of the diffusion layer, thus
`increasing the solubility of the parent compound, C,, in
`that layer over its inherent solubility at the pH of the
`dissolution medium. Hence, dissolution is controlled by
`solubility in the diffusion layer which, in turn, is deter-
`mined by the pH of that layer. The influence of Ksp on the
`solubility term, Cs, and dissolution rate, should an accu-
`mulation of ions be allowed to occur, will be treated
`later.
`Nelson (98), in a study of theophylline salts, was the first
`to show the correlation between diffusion layer pH and
`dissolution rate. The major impact that this study had on
`the pharmaceutical sciences was its conclusion that, if
`other factors remained constant, the dissolution rate of a
`compound determined the rate of buildup of blood levels
`with time and the maximum levels obtained. Those salts
`of the acidic theophylline with high diffusion layer pH’s
`had greater in uitro dissolution rates than those exhibiting
`a lower diffusion layer pH. And, indeed, the rank order of
`dissolution rates correlated well with clinically determined
`blood levels. Presumably, the higher pH in the diffusion
`layer retards hydrolysis of the salt, thereby maintaining
`the anionic charge of the theophyllinate ion. This report
`led to many additional studies which illustrate the influ-
`ence of the salt form on dissolution and the beneficial ef-
`fects of changing nonionized drugs into salts.
`Juncher and Raaschou (99) demonstrated that the rank
`order of peak blood levels of penicillin V, obtained upon
`
`’ ‘ h e autbws recognize the existence of other models; this one was chosen simply
`for illustwt ive purposes.
`
`6 1 Journal of Pharmaceutical Sciences
`
`administration of three different salts and the free acid,
`was the same as the rank order of their rates of dissolution
`in uitro. While the investigators ascribed these differences
`to the solubility properties of the salts, their experiments
`actually compared dissolution rates, not solubilities. The
`relative order of dissolution rates and mean maximal blood
`levels was: potassium salt > calcium salt > free acid >
`benzathine salt.
`Nelson (100) determined dissolution rates for several
`weak acids and their sodium salts in media whose pH’s
`represented GI fluids. In all cases, the sodium salt dis-
`solved more rapidly than the free acid. This finding re-
`solved the misconception that absorption of drugs is re-
`lated only to solubility in the appropriate medium; rather,
`solubility affects absorption only to the extent that it af-
`fects dissolution rate. Absorption of drugs is a dynamic
`process, and the ultimate solubility of a drug in fluid at
`absorption sites is of limited consequence since absorption
`prevents the attainment of saturated solutions. Therefore,
`dissolution rate, more than solubility, influences absorp-
`tion since it is a preceding process.
`In two subsequent studies, Nelson and coworkers fur-
`ther illustrated the effects of changing nonionized drugs
`into salts. A report concerning tolbutamide (101), a weak
`acid, showed that the initial dissolution rate of tolbutamide
`sodium was approximately 5000 times more rapid than the
`free acid in acidic media and 300 times more rapid in
`neutral media. This difference, measured in uitro, reflected
`the differences observed between the free a

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