throbber
modern Pharmaceutics
`
`Third Edition, Revised and Expanded
`
`edited by
`Gilbert S. Banker
`
`University of Iowa
`Iowa City, Iowa
`
`Christopher T. Rhodes
`
`University of Rhode Island
`Kingston, Rhode Island
`
`Marcel Dekker, Inc. (cid:9)
`
`New York• Basel • Hong Kong
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 1 of 35
`
`

`
`Library of Congress Cataloging-in-Publication Data
`
`Modern pharmaceutics / edited by Gilbert S. Banker, Christopher T.
`Rhodes.-3rd ed., rev. and expanded.
`p. cm.—(Drugs and the pharmaceutical sciences ; v. 72)
`Includes bibliographical references and index.
`ISBN 0-8247-9371-4 (alk. paper)
`1. Drugs—Dosage forms. 2. Biopharmaceutics.
`3. Pharmacokinetics. 4. Pharmaceutical industry—Quality control.
`I. Banker, Gilbert S. II. Rhodes, Christopher T. III. Series.
`RS200.M63 1995
`615'.1—dc20 (cid:9)
`
`95-33238
`CIP
`
`The publisher offers discounts on this book when ordered in bulk quantities. For more information, write
`to Special Sales/Professional Marketing at the address below.
`
`This book is printed on acid-free paper.
`
`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
`
`Current printing (last digit)
`10 9 8 7 6 5 4 3 2 1
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 2 of 35
`
`

`
`Chemical Kinetics and Drug Stability
`
`J. Keith Guillory and Rolland I. Poust
`College of Pharmacy, University of Iowa, Iowa City, Iowa
`
`6
`
`I. INTRODUCTION
`
`In the rational design and evaluation of dosage forms for drugs, the stability of the active
`components must be a major criterion in determining their suitability. Several forms of insta-
`bility can lead to the rejection of a drug product. First, there may be chemical degradation of
`the active drug, leading to a substantial lowering of the quantity of the therapeutic agent in
`the dosage form. Many drugs (e.g., digoxin and theophylline) have narrow therapeutic indices,
`and they need to be carefully titrated in individual patients so that serum levels are neither so
`high that they are potentially toxic, nor so low that they are ineffective. For these drugs, it is
`of paramount importance that the dosage form reproducibly deliver the same amount of drug.
`Second, although chemical degradation of the active drug may not be extensive, a toxic
`product may be formed in the decomposition process. Dearborn [I] described several examples
`in which the products of degradation are significantly more toxic than the original therapeutic
`agent. Thus, the conversions of tetracycline to epianhydrotetracycline, arsphenamine to oxo-
`phenarsine, and p-aminosalicylic acid to m-aminophenol in dosage forms give rise to potentially
`toxic agents that, when ingested, can cause undesirable effects. Recently, Nord et al. [2] re-
`ported that the antimalarial chloroquine can produce toxic reactions that are attributable to the
`photochemical degradation of the substance. Phototoxicity has also been reported to occur
`following administration of chlordiazepoxide and nitrazepam [3]. Another example of an ad-
`verse reaction caused by a degradation product was provided by Neftel et al. [4], who showed
`that infusion of degraded penicillin G led to sensitization of lymphocytes and formation of
`antipenicilloyl antibodies.
`Third, instability of a drug product can lead to a decrease in its bioavailability, rather than
`to loss of drug or to formation of toxic degradation products. This reduction in bioavailability
`can result in a substantial lowering in the therapeutic efficacy of the dosage form. This phe-
`nomenon can be caused by physical or chemical changes in the excipients in the dosage form,
`
`179
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 3 of 35
`
`

`
`180 (cid:9)
`
`Guillory and Poust
`
`independent of whatever changes the active drug may have undergone. A more detailed dis-
`cussion of this subject is given in Sec. II.B.
`Fourth, there may be substantial changes in the physical appearance of the dosage form.
`Examples of these physical changes include mottling of tablets, creaming of emulsions, and
`caking of suspensions. Although the therapeutic efficacy of the dosage form may be unaffected
`by these changes, the patient will most likely lose confidence in the drug product, which then
`has to be rejected.
`A drug product, therefore, must satisfy stability criteria chemically, toxicologically, thera-
`peutically, and physically. Basic principles in pharmaceutical kinetics can often be applied to
`anticipate and quantify the undesirable changes so that they can be circumvented by stabili-
`zation techniques. Some chemical compounds, called prodrugs [5,6], are designed to undergo
`chemical or enzymatic conversion in vivo to pharmacologically active drugs. Prodrugs are
`employed to solve one or several problems presented by active drugs (e.g., short biological
`half-life, poor dissolution, bitter taste, inability to penetrate through the blood—brain barrier,
`and others). They are pharmacologically inactive as such, but are converted back in vivo to
`their parent (active) compounds. Naturally, the rate and extent of this conversion (which are
`governed by the same laws of kinetics that will be described in this chapter) are the primary
`determinants of the therapeutic efficacy of these agents.
`In the present chapter, stability problems and chemical kinetics are introduced and surveyed.
`The sequence employed is as follows: first, an overview of the potential routes of degradation
`that drug molecules can undergo; then, a discussion of the mathematics used to quantify drug
`degradation; a delineation of the factors that can affect degradation rates, with an emphasis on
`stabilization techniques; and, finally, a description of stability-testing protocols employed in
`the pharmaceutical industry. It is not the intent of this chapter to document stability data of
`various individual drugs. Readers are referred to the compilations of stability data [7] and to
`literature on specific drugs [e.g., Ref. 8 and earlier volumes] for this kind of information.
`
`II. ROUTES BY WHICH PHARMACEUTICALS DEGRADE
`Since most drugs are organic molecules, it is important to recognize that many pharmaceutical
`degradation pathways are, in principle, similar to reactions described for organic compounds
`in standard organic chemistry textbooks. On the other hand, it is also important to realize that
`different emphases are placed on the types of reactions that are commonly encountered in the
`drug product stability area, as opposed to those seen in classic organic chemistry. In the latter,
`reactions are generally described as tools for use by the synthetic chemist; thus, the conditions
`under which they are carried out are likely to be somewhat drastic. Reactive agents (e.g., thionyl
`chloride or lithium aluminum hydride) are employed in relatively high concentrations (often >
`10%) and are treated using exaggerated conditions, such as refluxing or heating in a pressure
`bomb. Reactions are effected in relatively short time periods (hours or days). In contrast,
`reactions occurring in pharmaceuticals often involve the active drug components in relatively
`low concentrations. For example, dexamethasone sodium phosphate, a synthetic adrenocorti-
`coid steroid salt, is present only to the extent of about 0.4% in its injection, 0.1% in its topical
`cream or ophthalmic solution, and 0.05% in its ophthalmic ointment. The decomposition of a
`drug is likely to be mediated not by reaction with another active ingredient, but by reaction
`with water, oxygen, or light. Reaction conditions of interest are usually ambient or subambient.
`Reactions in pharmaceuticals ordinarily occur over months or years, as opposed to the hours
`or days required for completion of reactions in synthetic organic chemistry.
`Reactions such as the Diels-Alder reaction and aldol condensations, which are important in
`synthetic and mechanistic organic chemistry, are of only minor importance when drug degra-
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 4 of 35
`
`

`
`Chemical Kinetics and Drug Stability (cid:9)
`
`181
`
`dation is being considered. Students need to refocus their attention on reactions such as hy-
`drolysis, oxidation, photolysis, racemization, and decarboxylation, the routes by which most
`pharmaceuticals degrade.
`A cognizance of reactions of particular functional groups is important if one is to gain a
`broad view of drug degradation. It is a difficult task to recall degradative pathways of all
`commonly used drugs. Yet, through the application of functional group chemistry, it is possible
`to anticipate the potential mode(s) of degradation that drug molecules will likely undergo. In
`the following discussion, therefore, degradative routes are demonstrated by calling attention to
`the reactive functional groups present in drug molecules. The degradative routes are described,
`through the use of selected examples, as chemical when new chemical entities are formed as
`a result of drug decomposition, and as physical when drug, loss does not produce distinctly
`different chemical products.
`
`A. Chemical Degradative Routes
`
`Solvolysis
`In this type of reaction, the active drug undergoes decomposition following reaction with the
`solvent present. Usually, the solvent is water; but sometimes the reaction may involve phar-
`maceutical cosolvents, such as ethyl alcohol or polyethylene glycol. These solvents can act as
`nucleophiles, attacking the electropositive centers in drug molecules. The most common sol-
`volysis reactions encountered in pharmaceuticals are those involving "labile" carbonyl com-
`pounds, such as esters, lactones, and lactams (Table 1).
`Although all the functional groups cited are, in principle, subject to solvolysis, the rates at
`which they undergo this reaction may be vastly different. For example, the rate of hydrolysis
`of a 13-lactam ring (a cyclized amide) is much greater than that of its linear analog. The half-
`life (the time needed for half the drug to decompose) of the 13-lactam in potassium phenethi-
`cillin at 35°C and pH 1.5 is about 1 hr. The corresponding half-life for penicillin G is about
`4 min [9]. In contrast, the half-life for hydrolysis of the simple amide propionamide in 0.18
`molal 11,SO, at 25°C is about 58 hr [10]. It has been suggested that the antibacterial activity
`of p-lactam antibiotics arises from a combination of their chemical reactivity and their molec-
`ular recognition by target enzymes. One aspect of their chemical reactivity is their acylating
`power and, although penicillins are not very good acylating agents, they are more reactive than
`simple, unsubstituted amides [11]. Unactivated or "normal" amides undergo nonenzymatic
`hydrolysis slowly, except under the most extreme conditions of pH and temperature, because
`the N—C(0) linkage is inherently stable, yet when the amine function is a good leaving group
`(and particularly if it has a pK, greater than 4.5), amides can be susceptible to hydrolysis at
`ordinary temperatures. [For a recent review on this subject see Ref. 12.] Acyl-transfer reactions
`in peptides, including the transfer to water (hydrolysis), are of fundamental importance in
`biological systems in which the reactions proceed at normal temperatures, and enzymes serve
`as catalysts.
`The most frequently encountered hydrolysis reaction in drug instability is that of the ester,
`but certain esters can be stable for many years when properly formulated. Substituents can
`have a dramatic effect on reaction rates. For example, the tent-butyl ester of acetic acid is
`about 120 times more stable than the methyl ester, which, in turn, is approximately 60 times
`more stable than the vinyl analog [13]. Structure–reactivity relationships are dealt with in the
`discipline of physical organic chemistry. Substituent groups may exert electronic (inductive
`and resonance), steric, or hydrogen-bonding effects that can drastically affect the stability of
`compounds. Interested students are referred to a recent review by Hansch and Taft [14], and
`to the classic reference text written by Hammett [15].
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 5 of 35
`
`

`
`182 (cid:9)
`
`GulDory and Poust
`
`Table 1 Some Functional Groups Subject to Hydrolysis
`
`Drug type (cid:9)
`
`Esters
`
`RCOOR'
`
`RO PO
`3
`
`M
`
`x
`
`Examples
`
`Aspirin, alkaloids
`Dexamethasone sodium phosphate
`Estrone sulfate
`Nitroglycerin
`
`Lactones
`
`R
`
`Amides
`
`Lactams
`
`Oximes
`
`Imides
`
`Malonic ureas
`
`R
`
`ROS0
`
`3
`RO NO
`
`M
`
`x
`
`2
`
`0
`
`RCONR'
`
`C = NOR
`
`R
`
`2
`
`C=0
`NH
`C=0
`0
`II
`C—NH
`C=0
`C—NH
`II
`0
`
`Pilocarpine
`Spironolactone
`
`Thiacinamide
`Chloramphenicol
`
`Penicillins
`Cephalosporins
`
`Steroid oximes
`
`Glutethimide
`Ethosuximide
`
`Barbiturates
`
`Nitrogen mustards
`
`/
`R-N
`
`
`
`CH2CH2C1
`
`CH2CH2C1
`
`Melphalan
`
`A dramatic decrease in ester stability can be brought about by intramolecular catalysis. This
`type of facilitation is affected mostly by neighboring groups capable of exhibiting acid—base
`properties (e.g., —NH2, —OH, —COOH, and COO—). If neighboring-group participation leads
`to an enhanced reaction rate, the group is said to provide anchimeric assistance [16]. For
`example, the ethyl salicylate anion undergoes hydrolysis in alkaline solution at a rate that is
`106 times larger than the experimental value for the uncatalyzed cleavage of ethyl p-hydroxy-
`benzoate. The rate advantage is attributed to intramolecular general base catalysis by the phen-
`olate anion [17].
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 6 of 35
`
`(cid:9)
`(cid:9)
`

`
`Chemical Kinetics and Drug Stability (cid:9)
`
`183
`
`Oxidation
`Oxidation reactions are important pathways of drug decomposition. In pharmaceutical dosage
`forms, oxidation is usually mediated through reaction with atmospheric oxygen under ambient
`conditions, a process commonly referred to as autoxidation. Oxygen is, itself, a diradical, and
`most autoxidations are free-radical reactions. A free radical is a molecule or atom with one or
`more unpaired electrons. Of considerable importance to pharmaceutical scientists is a reliable
`method for determining and controlling oxygen concentration in aqueous solutions [18]. A
`thorough review of autoxidation and of antioxidants has been published [19].
`The mechanisms of oxidation reactions are usually complex, involving multiple pathways
`for the initiation, propagation, branching, and termination steps. Many autoxidation reactions
`are initiated by trace amounts of impurities, such as metal ions or hydroperoxides. Thus, ferric
`ion catalyzes the degradation reaction and decreases the induction period for the oxidation of
`the compound procaterol [20]. As little as 0.0002 M copper ion will increase the rate of vitamin
`C oxidation by a factor of 105 [21]. Hydroperoxides contained in polyethylene glycol suppos-
`itory bases have been implicated in the oxidation of codeine to codeine-N-oxide [22]. Peroxides
`apparently are responsible for the accelerated degradation of benzocaine hydrochloride in aque-
`ous cetomacrogol solution [23] and of a corticosteroid in polyethylene glycol 300 [24,25].
`Many oxidation reactions are catalyzed by acids and bases [26].
`A list of some functional groups that are subject to autoxidation is shown in Table 2. The
`products of oxidation are usually electronically more conjugated; thus, the appearance of, or a
`change in, color in a dosage form is suggestive of the occurrence of oxidative degradation.
`
`Photolysis
`Normal sunlight or room light may cause substantial degradation of drug molecules. The energy
`from light radiation must be absorbed by the molecules to cause a photolytic reaction. If that
`
`Table 2 Some Functional Groups Subject to Autoxidation
`
`Functional group (cid:9)
`
`Phenols
`
`Catechols
`
`Ethers
`
`Thiols
`
`Thioethers
`
`Carboxylic acids
`
`Nitrites
`
`Aldehydes
`
`HO
`
`HO
`
`HO
`
`R —O —R'
`
`RCH
`
`SH
`
`2
`
`R —S — R'
`
`RCOOH
`
`R NO 2
`
`RCHO
`
`Examples
`
`Phenols in steroids
`
`Catecholamines (dopamine,
`isoproterenol)
`
`Diethylether
`
`Dimercaprol (BAL)
`
`Phenothiazines (chlorpromazine)
`
`Fatty acids
`
`Amyl nitrite
`
`Paraldehyde
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 7 of 35
`
`

`
`184 (cid:9)
`
`Guillory and Poust
`
`energy is sufficient to achieve activation, degradation of the molecule is possible. Saturated
`molecules do not interact with visible or near-ultraviolet light, but molecules that contain ar-
`electrons usually do absorb light throughout this wavelength range. Consequently, compounds
`such as aromatic hydrocarbons, their heterocyclic analogues, aldehydes, and ketones, are most
`susceptible to photolysis. In general, drugs that absorb light at wavelengths below 280 nm
`have the potential to undergo decomposition in sunlight, and drugs with absorption maxima
`greater than 400 nm have the potential for degradation both in sunlight and room light.
`A dramatic example of photolysis is the photodegradation of sodium nitroprusside in aque-
`ous solution. Sodium nitroprusside, Na2Fe(CN)5N0.21-120, is administered by intravenous
`infusion for the management of acute hypertension. If the solution is protected from light, it
`is stable for at least 1 year; if exposed to normal room light, it has a shelf life of only 4 hr
`[27].
`Photolysis reactions are often associated with oxidation because the latter category of re-
`actions can frequently be initiated by light. But, photolysis reactions are not restricted to oxi-
`dation. For sodium nitroprusside, it is believed that degradation results from loss of the nitro-
`ligand from the molecule, followed by electronic rearrangement and hydration. Photo-induced
`reactions are common in steroids [28]; an example is the formation of 2-benzoylcholestan-3-
`one following irradiation of cholest-2-en-3-ol benzoate. Photoadditions of water and of alcohols
`to the electronically excited state of steroids have also been observed [29].
`
`Dehydration
`The preferred route of degradation for prostaglandin E2 and tetracycline is the elimination of
`a water molecule from their structures. The driving force for this type of covalent dehydration
`is the formation of a double bond that can then participate in electronic resonance with neigh-
`boring functional groups. In physical dehydration processes, such as those occurring in the-
`ophylline hydrate and ampicillin trihydrate [30], water removal does not create new bonds, but
`often changes the crystalline structure of the drug. Since it is possible that anhydrous com-
`pounds may have different dissolution rates compared with their hydrates [31,32], dehydration
`reactions involving water of crystallization may potentially affect the absorption rates of the
`dosage form.
`
`Racemization
`The racemization of pharmacologically active agents is of interest because enantiomers often
`have significantly different absorption, distribution, metabolism, and excretion, in addition to
`differing pharmacological actions [33]. The best-known racemization reactions of drugs are
`those that involve epinephrine, pilocarpine, ergotamine, and tetracycline. In these drugs, the
`reaction mechanism appears to involve an intermediate carbonium ion or carbanion that is
`stabilized electronically by the neighboring substituent group. For example, in the racemization
`of pilocarpine [34], a carbanion is produced and stabilized by delocalization to the enolate. In
`addition to the racemization reaction, pilocarpine is also degraded through hydrolysis of the
`lactone ring.
`Most racemization reactions are catalyzed by an acid or by a base. A notable exception is
`the "spontaneous" racemization of the diuretic and antihypertensive agent, chlorthalidone,
`which undergoes facile SN1 solvolysis of its tertiary hydroxyl group to form a planar carbonium
`ion. Chiral configuration is then restored by nucleophilic attack (S,2) of a molecule of water
`on the carbonium ion, with subsequent elimination of a proton [35].
`
`Incompatibilities
`Chemical interactions between two or more drug components in the same dosage form, or
`between active ingredient and a pharmaceutical adjuvant, frequently occur. An example of
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 8 of 35
`
`

`
`Chemical Kinetics and Drug Stability (cid:9)
`
`185
`
`drug—drug incompatibility is the inactivation of cationic aminoglycoside antibiotics, such as
`kanamycin and gentamicin, by anionic penicillins in IV admixtures. The formation of an in-
`active complex between these two classes of antibiotics occurs not only in vitro, but apparently
`also in vivo in patients with severe renal failure [36]. Thus, when gentamicin sulfate was given
`alone to patients on long-term hemodialysis, the biological half-life of gentamicin was greater
`than 60 hr. But, when carbenicillin disodium (CD) was given with gentamicin sulfate (GS) in
`the dose ratio CD/GS = 80:1, the gentamicin half-life was reduced to about 24 hr.
`Many pharmaceutical incompatibilities are the result of reactions involving the amine func-
`tional group. A summary of the potential interactions that can occur between various functional
`groups is given in Table 3.
`
`Other Chemical Degradation Reactions
`Other chemical reactions, such as hydration, decarboxylation, or pyrolysis, also are potential
`routes for drug degradation. Thus, cyanocobalamin may absorb about 12% of water when
`exposed to air, and p-aminosalicylic acid decomposes with evolution of carbon dioxide to form
`m-aminophenol when subjected to temperatures above 40°C. The temperature at which pyro-
`lytic decomposition of terfenadine occurs has been used as a criterion for determining which
`of several tablet excipients will be perferable for long-term stability of the drug substance [37].
`
`B. Physical Degradative Routes
`
`Polymorphs are different crystal forms of the same compound [38]. They are usually prepared
`by crystallization of the drug from different solvents under diverse conditions. Steroids, sul-
`fonamides, and barbiturates are notorious for their propensity to form polymorphs [39]. Yang
`and Guillory [40] attempted to correlate the occurrence frequency of polymorphism in sulfon-
`amides with certain aspects of chemical structure. They found that sulfonamides that did not
`exhibit polymorphism have somewhat higher melting points and heats of fusion than those that
`were polymorphic. The absence of polymorphism in sulfacetamide was attributed to the
`stronger hydrogen bonds formed by the amide hydrogen in this molecule. These stronger
`hydrogen bonds were not readily stretched or broken to form alternate crystalline structures.
`Since polymorphs differ from one another in their crystal energies, the more energetic ones
`will seek to revert to the most stable (and the least energetic) crystal form. When several
`polymorphs and solvates (substances that incorporate solvent in a stoichiometric fashion into
`the crystal lattice) are present, the conditions under which they may interconvert can become
`quite complex, as is true of fluprednisolone [41].
`Polymorphs may exhibit significant differences in important physicochemical parameters,
`such as solubility, dissolution rate, and melting point [42]. Thus, the conversion from one
`polymorph to another in a pharmaceutical dosage form may lead to a drastic change in the
`physical characteristics of the drug. A well-known example of this phenomenon is the con-
`version of a more soluble crystal form (form II) of cortisone acetate to a less soluble form
`(form V) when the drug is formulated into an aqueous suspension [43]. This phase change
`leads to caking of the cortisone acetate suspension.
`Another physical property that can affect the appearance, bioavailability, and chemical sta-
`bility of pharmaceuticals is the degree of crystallinity. It has been reported that crystalline
`insulin [44] and crystalline cyclophosphamide [45] are much more stable than their amorphous
`counterparts.
`
`Vaporization
`Some drugs and pharmaceutical adjuvants possess sufficiently high vapor pressures at room
`temperature that their volatilization through the container constitutes a major route of drug
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 9 of 35
`
`

`
`186 (cid:9)
`
`Guillory and Poust
`
`Table 3 Some Potential Drug Incompatibilities
`
`Schiff's base formation
`
`(e g , aminophylline + glucose
`penicillamine + acetaldehyde)
`
`Racemization
`
`(e.g., epinephrine + HSO3)
`
`Amine (cid:9)
`
`Precipitation
`
`(e.g., epinephrine sulfate + steroid
`phosphate sodium)
`
`1. Amide formation
`(e.g., benzocaine + citric acid)
`2. Salt/complex formation
`(e.g., gentamicin + carbenicillin)
`
`Aminolysis
`
`(e.g., aspirin + phenylephrine)
`
`Aldehyde
`
`Bisulfite adduct
`
`Bisulfite
`
`Large anions
`
`Carboxylic acid
`
`Ester
`
`Alcoholysis
`(e.g., aspirin + PEG)
`
`L Alcohol
`
`loss. Flavors, whose constituents are mainly ketones, aldehydes and esters, and cosolvents (low
`molecular weight alcohols) may be lost from the formulation in this manner. The most fre-
`quently cited example of a pharmaceutical that "degrades" by this route is nitroglycerin, which
`has a vapor pressure of 0.00026 mm at 20°C and 0.31 mm at 93°C [46]. Significant drug loss
`to the environment can occur during patient storage and use. In 1972, the Food and Drug
`Administration (FDA) issued special regulations governing the types of containers that may
`be used for dispensing sublingual nitroglycerin tablets [47].
`Reduction of vapor pressure, and thereby of volatility, of drugs such as nitroglycerin can
`be achieved through dispersion of the volatile drug in macromolecules that can provide phys-
`icochemical interactions. The addition of macromolecules, such as polyethylene glycol, poly-
`vinylpyrrolidone, and microcrystalline cellulose, allows preparation of "stabilized" nitroglyc-
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 10 of 35
`
`

`
`Chemical Kinetics and Drug Stability (cid:9)
`
`187
`
`erin sublingual tablets [48,49]. A 13-cyclodextrin—nitroglycerin tablet is currently being
`marketed in Japan to achieve the same purpose.
`Another aspect of nitroglycerin instability has been observed by Fusari [49]. When conven-
`tional (unstabilized) nitroglycerin sublingual tablets are stored in enclosed glass containers, the
`high volatility of the drug gives rise to redistribution of nitroglycerin among the stored tablets.
`Interestingly, this redistribution leads to an increase in the standard deviation of the drug
`contents of the tablets, rather than the reverse. This migration phenomenon results in a dete-
`rioration in the uniformity of the tablets on storage.
`
`Aging
`The most interesting, and perhaps the least-reported, area of concern about the physical insta-
`bility of pharmaceutical dosage forms is generally termed aging. This is a process through
`which changes in the disintegration or dissolution characteristics of the dosage form are caused
`by subtle, and sometimes unexplained, alterations in the physicochemical properties of the inert
`ingredients or the active drug in the dosage form [50]. Since the disintegration and dissolution
`steps may be the rate-determining steps in the absorption of a drug, changes in these processes,
`as a function of the "age" of the dosage form, may result in corresponding changes in the
`bioavailability of the drug product.
`An example of this phenomenon was provided by deBlaey and Rutten-Kingma [51], who
`showed that the melting time of aminophylline suppositories, prepared from various bases,
`increased from about 20 min to over an hour after 24 weeks of storage at 22°C. Like the
`dissolution time for solid dosage forms, the melting time for suppositories can be viewed as
`an in vitro index of drug release. Thus, an increase in melting time can perceivably lead to a
`decrease in bioavailability. The mechanism responsible for this change appeared to involve an
`interaction between the ethylenediamine in aminophylline and the free fatty acids present in
`the suppository bases. Interestingly, no increase in melting time was detected when the sup-
`positories were stored at 4°C, even up to 15 months.
`Aging of solid dosage forms can cause a decrease in their in vitro rate of dissolution [52],
`but a corresponding decrease in in vivo absorption cannot be assumed automatically. For ex-
`ample, Chemburkar et al. [53] showed that, when a methaqualone tablet was stored at 80%
`relative humidity for 7-8 months, the dissolution rate, as measured by in vivo absorption, was
`not affected. This lack of in vitro dissolution—in vivo absorption correlation for the aged
`product was observed even through the particular dissolution method (that of the resin flask)
`was shown by the same workers to be capable of discriminating the absorption of several trial
`dosage forms of the same drug.
`
`Adsorption
`Drug—plastic interaction is increasingly being recognized as a major potential problem when
`intravenous solutions are stored in bags, or when they are infused through administration sets
`that are made from polyvinyl chloride (PVC). For example, up to 50% drug loss can occur
`after nitroglycerin is stored in PVC infusion bags for 7 days at room temperature [54]. This
`loss can be attributed to adsorption, rather than to chemical degradation, because the drug can
`be recovered from the inner surface of the container by rinsing with a less polar solvent
`(methanol here). A diverse array of drugs, including diazepam [55], insulin [56], isosorbide
`dinitrate [57], and others [58], have shown substantial adsorption to PVC. The propensity for
`significant adsorption is related to the oil/water partition coefficient of the drug, since this
`process depends on the relative affinity of the drug for the hydrophobic PVC (dielectric constant
`of about 3) and the hydrophilic aqueous infusion medium.
`
`NOVARTIS EXHIBIT 2014
`Noven v. Novartis and LTS Lohmann
`IPR2014-00550
`Page 11 of 35
`
`

`
`188 (cid:9)
`
`Guillory and Poust
`
`Physical Instability in Heterogeneous Systems
`The stability of suspensions, emulsions, creams, and ointments is dealt with in other chapters.
`The unique characteristics of solid-state decomposition processes have been described in re-
`views by Monkhouse [59,60] and in the more recently published monograph on drug stability
`by Carstensen [61].
`
`III. QUANTITATION OF RATE OF DEGRADATION
`
`Before undertaking a discussion of the mathematics involved in the determination of reaction
`rates is undertaken, it is necessary to point out the importance of proper data acquisition in
`stability testing. Applications of rate equations and predictions are meaningful only if the data
`used in such processes are collected using valid statistical and analytical procedures. It is
`beyond the scope of this chapter to discuss the proper statistical treatments and analytical
`techniques that should be used in a stability study. But, some perspectives in these areas can
`be obtained by reading the comprehensive review by Meites [62] and from the section on
`statistical considerations in the stability guidelines published by the FDA in 1987 [63].
`
`A. Kinetic Equations
`
`Consider the reaction
`
`aA + bB --> mM + nN
`
`(1)
`
`where A and B are the reactants; M and N, the products; and a, b, m, and n, the stoichiometric
`coefficients describing the reaction. The rate of change of the concentration C of any of the
`species can be expressed by the differential notations —dCA/dt, —dCB/dt, dCwildt, and dCadt.
`Note that the rates of change for the reactants are preceded by a negative sign, denoting a
`decrease in concentration relative to time (rate of disappearance). In contrast, the differential
`terms for the products are positive in sign, indicating an increase in concentration of these
`species as time increases (rate of appearance). The rates of disappearance of A and B and the
`rates of appearance of M and N are interrelated by equations that take into account the stoi-
`chiometry of the reaction:
`
`1 dCA (cid:9)
`1 dCN
`1 dCB 1 (cid:9)
`a dt - b dt - m dt - n dt
`
`(2)
`
`The Rate Expression
`The rate expression is a mathematical description of the rate of the reaction at any time t in
`terms of the concentrat

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