throbber
REVIEW
`
`Excipient–Drug Interactions in Parenteral Formulations
`
`MICHAEL J. AKERS
`
`Baxter Healthcare Corporation, Bloomington, Indiana 47402
`
`Received 10 December 2001; revised 12 February 2002; accepted 18 February 2002
`
`ABSTRACT: Excipients are added to parenteral formulations to enhance or maintain
`active ingredient solubility (solubilizers) and/or stability (buffers, antioxidants, chelat-
`ing agents, cryo- and lyoprotectants). Excipients also are important in parenteral formu-
`lations to assure safety (antimicrobial preservatives), minimize pain and irritation upon
`injection (tonicity agents), and control or prolong drug delivery (polymers). These are all
`examples of positive or synergistic interactions between excipients and drugs. However,
`excipients may also produce negative effects such as loss of drug solubility, activity, and/
`or stability. This review article will highlight documented interactions, both synergistic
`and antagonistic, between excipients and drugs in parenteral formulations. The reader
`will gain better understanding and appreciation of the implications of adding formu-
`lation ingredients to parenteral drug products. ß 2002 Wiley-Liss, Inc. and the American
`Pharmaceutical Association J Pharm Sci 91:2283–2300, 2002
`Keywords: parenteral; excipients; formulation; stabilizers; solubilizers; antimicro-
`bial preservatives; packaging
`
`INTRODUCTION
`
`Well-referenced and useful publications are avail-
`able listing every formulation component in all
`marketed parenteral drug products1–5 (Food and
`Drug Administration web sitea). The information
`in these publications has been invaluable to
`parenteral formulation scientists developing solu-
`ble, stable, resuspendable, manufacturable, and
`deliverable parenteral dosage forms. Formulation
`component precedence takes on high stature in
`the sterile product world because of significant
`toxicological and regulatory concerns. In other
`words, it is usually better to use a component
`that has a track record of relatively safe use in
`injectables and is likely not to raise concerns on
`the part of regulatory reviewers.
`
`awww.fda.gov/cder/drug/iig/default.htm
`
`Correspondence to: Michael J. Akers (Telephone: 812-355-
`7188; Fax: 812-332-3079; E-mail: michael_akers@baxter.com)
`
`Journal of Pharmaceutical Sciences, Vol. 91, 2283–2300 (2002)
`ß 2002 Wiley-Liss, Inc. and the American Pharmaceutical Association
`
`This review article will not cover all excipients
`used in parenteral formulations because the afore-
`mentioned publications already do so. What this
`review article presents are examples of synergistic
`and antagonist interactions that have been report-
`ed for excipients used in parenteral formulations.
`Although extensive, this review will not be exhaus-
`tive in the effort to cite all published references on
`parenteral drug–excipient interactions. Pharma-
`ceutical Excipients 20006 was a very helpful text in
`obtaining valuable information about drug–exci-
`pient interactions and compatibilities.
`When one studies stability and compatibility
`issues in parenteral drug formulation, the packa-
`ging system also must be considered. Potential
`interactions between excipients and rubber clo-
`sures in finished products are as much a concern
`as interactions between excipients and drugs.
`Therefore, some drug–sterile packaging interac-
`tions will be covered in this article.
`Although this article will focus on chemical and
`physical compatibilities of drugs and excipients
`used as injectable products, readers must also be
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
`2283
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 1 of 18
`
`

`

`2284
`
`AKERS
`
`aware of the potential for any excipient and drug,
`either alone or in combination, when injected
`intravenously to cause certain problems. As
`Yalkowsky et al.7 pointed out, some formulation-
`related problems associated with intravenous
`drug delivery include hemolysis, precipitation,
`phlebitis, and pain. Therefore, as scientists de-
`velop sterile product formulations, not only must
`they be concerned with physical and chemical in-
`teractions that may occur in vitro, but they must
`also be concerned with the potential for formula-
`tion-related problems occurring in vivo.
`Drug–excipient interactions are studied in two
`basic ways. One is to perform traditional prefor-
`mulation studies using full factorial or Plackett
`Burman type of experimental designs. A good
`example of this approach for parenteral formu-
`lation development is a preformulation study
`published by Peswani and Lalla.8 Analytical
`methods such as differential scanning calorimetry
`isothermal microcalorimetry,10,11 and
`(DSC),9
`fourier transform infrared (FT-IR) spectroscopy12
`are excellent tools for predicting drug–excipient
`interactions. The other approach for studying
`drug–excipient interactions is to conduct both
`short-term and long-term stability studies on
`various formulations of the drug and measure
`both chemical stability (usually by chromatogra-
`phic techniques) and physical stability (e.g., by
`microscopic, electronic particle analysis, and cir-
`cular dichroism techniques).
`This review is organized according to major
`functions of parenteral excipients (solubilization,
`stabilization, preservation, and drug delivery aids).
`Several excipients serve more than one function
`[e.g., polyvinylpyrrolidone (PVP) as a complexing
`agent and as a freeze-drying bulking agent], so
`such excipients may be referenced in more than
`one segment of the article.
`Table 1 lists all the major pharmaceutical ex-
`cipients used in parenteral formulations. Table 2
`provides a listing of lesser-used excipients that
`are found in 1–2 commercial parenteral formula-
`tions. References1–5 provide much greater detail
`about the specifics of these excipients (e.g., con-
`centration) and the products in which they are
`components (e.g., brand names, manufacturer).
`
`Solubility Effects
`
`Many parenteral formulations require additives,
`either solvent or solute excipient, to increase and/
`or maintain solubility of the active ingredient in
`the solution. Sweetana and Akers13 summarized
`
`seven basic approaches for solubilization of paren-
`teral drugs as follows:
`
`1. Salt formation
`2. pH adjustment
`3. Use of co-solvents
`4. Use of surface-active agents
`5. Use of complexation agents
`6. Change formulation from solution to a
`dispersed system, oily solution, or a more
`complex formulation such as a microemul-
`sion or liposome
`7. ‘‘Heroic’’ approaches involving the use ofnon-
`commercially approved types and/or concen-
`trations of solvents or excipients
`
`This section will highlight some of the interac-
`tions between drugs and solubilizing agents,
`focusing on co-solvents, surfactants, suspending
`and emulsifying agents, complexation agents, and
`oils or lipids. Some examples of unpredicted inter-
`actions of excipients and drugs to enhance drug
`solubility are listed in Table 3.
`
`Co-Solvents
`
`There are approximately 20 different co-solvent
`agents used in approved parenteral products.
`However, the most commonly used co-solvents in
`parenteral formulations are ethanol, glycerin, pro-
`pylene glycol, sorbitol, polyethylene glycol (both
`300 and 400), dimethylacetamide, Cremophor EL,
`and N-methyl-2-pyrrolidone.
`Glycols are widely used solubilizing agents,
`but can cause some stability or compatibility
`problems. Glycerol is used not only as a co-solvent
`for improving solubility of poorly water-soluble
`drugs, but also as a tonicity-adjusting agent (e.g.,
`in insulin formulations). In freeze-dried formula-
`tions, glycerol can serve as a plasticizer, lowering
`the glass transition temperature of the product
`without the significant change in water content or
`activity.14 In certain formulations containing un-
`stable peptides, the presence of glycerol will in-
`crease the mobility of the freeze-dried formulation
`matrix, leading to peptide deamidation. Sorbitol
`has been reported to increase the degradation
`rate of penicillins in neutral and aqueous solu-
`tions.15 On a more positive note, propylene glycol
`will potentiate the antimicrobial activity of the
`parabens in the presence of nonionic surfactants
`and prevents the interaction of methylparaben
`and polysorbate 80.16
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 2 of 18
`
`

`

`Table 1. A Listing of Major Excipients Used in
`Sterile Product Formulations (Both Commercial and
`Developmental)
`
`Solvent systems
`Co-solvents
`Propylene glycol
`Glycerin
`Ethanol
`Polyethylene glycol (300 and 400)
`Sorbitol
`Dimethylacetamide
`Cremophor EL
`Oils
`Sesame
`Soybean
`Corn
`Castor
`Cottonseed
`Peanut
`Arachis
`Ethyl oleate
`Isopropyl myristate
`Glycofurol
`Petrolatum
`Solubilization agents
`Co-solvents
`See above
`Surface-active agents
`Polyoxyethylene sorbitan monooleate (Tween 80)
`Sorbitan monooleate
`Polyoxyethylene sorbitan monolaurate (Tween 20)
`Lecithin
`Polyoxyethylene–polyoxypropylene copolymers
`(Pluronics1)
`Complexation agents
`Hydroxypropyl-b-cyclodextrin
`Sulfobutylether-b-cyclodextrin (Captisol1)
`Polyvinylpyrrolidone
`Amino acids (arginine, lysine, histidine)
`Stabilization agents
`Buffers
`Acetate
`Citrate
`Tartrate
`Phosphate
`Triethanolamine (TRIS)
`Antioxidants
`Ascorbic acid
`Acetylcysteine
`Sulfurous acid salts (bisulfite, metabisulfite)
`Monothioglyercol
`Chelating agents
`Ethylenediaminetetraacetic acid (EDTA)
`Sodium citrate
`Cryo- and lyoprotectants and bulking agents
`Mannitol
`Glycine
`Sucrose
`
`EXCIPIENT–DRUG INTERACTIONS
`
`2285
`
`Table 1.
`
`(Continued )
`
`Lactose
`Trehalose
`Dextran
`Povidone
`Sorbitol
`Competitive binding agents
`Serum albumin
`Heta-starch
`Tonicity-adjusting agents
`Sodium chloride
`Glycerin
`Mannitol
`Dextrose
`Antimicrobial preservative agents
`Phenol
`Meta-cresol
`Benzyl alcohol
`Parabens (methyl, propyl, butyl)
`Benzalkonium chloride
`Chlorobutanol
`Thimerosal
`Phenylmercuric salts (acetate, borate, nitrate)
`Delivery polymers
`See Table 6
`
`Co-solvents are known to cause hemolysis. In
`a study conducted by Fuet et al.17 comparing the
`hemolytic effects, both in vitro and in vivo,
`of a variety of co-solvents (ethanol, propylene gly-
`col, polyethylene glycol, dimethylisosorbide, and
`dimethylacetamide), complexing agents (nicotina-
`mide), and surfactants (Pluronic L64 and emul-
`phor EL-719), solutions most prone to elicit a
`hemolytic response were those containing propy-
`lene glycol, dimethylisosorbide, and nicotina-
`mide). However, these authors found that the
`hemolytic effects of propylene glycol can be alle-
`viated by the addition of either a tonicifying agent
`or polyethylene glycol 400.
`Cremophor EL (polyoxyl 35 castor oil) has
`been approved as a solvent in commercial in-
`jectable dosage forms containing paclitaxel,
`diazepam, propanidid, and alfaxalone. It is com-
`patible with many organic solvents and aqu-
`eous solutions. However, compounds containing
`phenolic hydroxyl groups may cause precipita-
`tion of Cremophor EL.
`
`Surfactants
`
`Surfactants serve a variety of very important
`functions in parenteral formulations. Among the
`most important are stabilizing proteins against
`aggregation. Tween 20 (polyoxyethylene sorbitan
`monolaurate) was shown to greatly reduce the
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 3 of 18
`
`

`

`2286
`
`AKERS
`
`Table 2. Examples of Special or Uncommon Excipients Used in Injectable Drug
`Products
`
`Excipient
`
`Product
`
`Manufacturer
`
`Acacia
`Acetone sodium
`Aluminum monostearate
`Benzenesulfonic acid
`Benzyl benzoate
`Cyclodextrin (alpha)
`Diethanolamine
`Desoxycholate sodium
`Formaldehyde
`
`Tuberculin Old Test (ID)
`Talwin (IM)
`Solganal
`Tracrium
`Depo-testesterone
`Alprostadil
`Bactrim
`Fungizone
`Some vaccines
`
`Gelatin, hydrolyzed
`Gelatin, purified
`Hydroxypropyl-b-cyclodextrin
`Imidazole
`Monoethanolamine
`N,N-dimethylacetamide
`
`Polyoxyethylated fatty acid
`PEG 40 castor oil
`PEG 60 castor oil
`Sodium lauryl sulfate
`Sulfobutylether-b-cyclodextrin
`Triacetin
`
`Some vaccines
`Lupron Depot
`Itraconazole
`Kogenate
`Terramycin (IM)
`Vuman
`Busulfan
`AquaMephyton
`Monistat
`Prograf
`Proleukin
`Ziprasidone mesylate
`Prepidil Gel (ICV)
`
`Lederle
`Sanofi Winthrop
`Schering
`Glaxo Smith Kline
`Pharmacia
`Schwarz
`Roche
`Bristol Myers Squibb
`Lederle, Connaught,
`Merck
`Merck
`TAP
`Janssen
`Bayer
`Roerig
`Bristol Myers Squibb
`Orphan
`Merck
`Janssen
`Fujisawa
`Cetus
`Pfizer
`Pharmacia
`
`rate of formation of insoluble aggregates of re-
`combinant human factor XIII caused by both
`freeze thawing and agitation stresses18 (Fig. 1).
`Maximum protection occurs at concentrations
`close to the critical micelle concentration of
`Tween 20, independent of initial protein concen-
`tration. In another report, Tween 20 at a 1% (w/v)
`concentration caused precipitation of a relatively
`hydrophobic protein (Humicola lanuginosa lipase)
`by inducing non-native aggregates.19
`Tween 80 is well known to protect proteins
`against surface-induced denaturation.20 Tween 80
`was demonstrated to reduce hemoglobin aggre-
`gation in solution by preventing the protein from
`reaching the air–liquid interface or the liquid–
`surface interfaces.21 Polyoxyethylene surfactants
`
`such as Tween 80 can form peroxide impurities
`after long-term storage. Knepp et al.22 concluded
`that Tween 80 and other nonionic polyether sur-
`factants undergo oxidation during bulk material
`storage and subsequent use and the resultant
`alkyl hydroperoxides formed can contribute to
`the degradation of proteins. In such formulations,
`they further reported that thiols such as cysteine,
`glutathione, and thioglycerol were most effective
`in stabilizing protein formulations containing
`peroxide-forming nonionic surfactants.
`The nonionic surfactant octoxynol 40 (ethoxy-
`lated alkyl phenol, Igepal CA897, GAF), was found
`to solubilize an otherwise insoluble complex of a
`nonsteroidal anti-inflammatory drug and a quar-
`ternary ammonium antimicrobial preservative
`
`Table 3. Examples of Esoteric Excipients Used as Solubilizers
`
`Generic Name
`
`Brand Name
`
`Manufacturer
`
`Excipient
`
`Cipro IV
`Ciprofloxacin
`Adriamycin RDF
`Doxorubicin HCl
`Ergonovine maleate
`Ergotrate Maleate
`Polyestradiol phosphate Estradurin
`Zomepirac
`Zomax
`
`Bayer
`Pharmacia
`Lilly
`Wyeth
`McNeil
`
`Lactic acid
`Methyl paraben
`Ethyl lactate
`Niacinamide
`Tromethamine105
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 4 of 18
`
`

`

`EXCIPIENT–DRUG INTERACTIONS
`
`2287
`
`products worldwide that contain cyclodextrins
`(see Table 2). However, based on the literature
`and scientific meeting presentations, there will be
`a higher number of
`cyclodextrin-containing
`injectable formulations in the future.
`The only reports of incompatibilities with cy-
`clodextrins involve certain antimicrobial preser-
`vatives, primarily parabens.26,27 One preliminary
`report described both sulfobutylether-b-cyclodex-
`trin and hydroxylpropyl-b-cyclodextrin accelerat-
`ing the degradation of an unidentified water-
`soluble drug to its insoluble degradant form.28
`The authors concluded that both the type and
`degree of substitution of the proximal hydroxyl
`groups in the cyclodextrin cavity will influence
`the potential for cyclodextrin additives to accel-
`erate chemical degradation of drugs. As cyclodex-
`trins become more prominent in injectable drug
`product development, there likely will be more
`reports of incompatibilities along with the ex-
`pected reports describing solubility and stability
`enhancements.
`Cyclodextrin-containing formulations (either
`0.1 M sulfobutylether-b-cyclodextrin or 0.1 M
`hydroxylpropyl-b-cyclodextrin) were shown to
`cause less damage to venous epithelial cells at
`the site of injection compared with formulations
`containing organic co-solvents.29 PVP (povidone)
`is a generally compatible polymeric excipient.
`However, it can form molecular adducts (a posi-
`tive reaction with respect to iodine therapy
`topically) and will complex with some preserva-
`tives such as thimerosal.6 Lecithin is a commonly
`used emulsifying and stabilizing agent in intra-
`muscular and intravenous injections, primarily
`the intravenous fatty or lipid emulsions used in
`parenteral nutrition. Lecithin also is a component
`of some liposomal formulations. Polaxamers (e.g.,
`Poloxamer 188, BP) are nonionic polyoxyethy-
`lene–polyoxypropylene copolymers used as emul-
`sifying agents in intravenous fat emulsions. They
`have also been used in several patented protein
`formulations as stabilizers and sustained release
`injectables in development as solubilizing and
`stabilizing agents.30 Polaxamers, like the poly-
`sorbates, can form peroxide impurities over time
`and are incompatible with antimicrobial preser-
`vatives such as phenol and paraben.
`
`Oils/Lipids
`
`Many commercially available parenteral products
`contain lipophilic or oleaginous solvents. Exam-
`ples of
`injectable lipid solvents include ethyl
`
`Figure 1. Recovery of native rFXIII (A) and forma-
`tion of soluble (B) and insoluble (C) aggregates after 10
`freeze–thaw cycles of 1 mg/mL (*), 5 mg/mL (*), and
`10 mg/mL (!) as a function of Tween 20. (From
`Krielgaard et al., J Pharm Sci, 87, 1593–1603, ß 1998
`John Wiley & Sons, Inc., reproduced with permission.)
`
`mixture in an ophthalmic formulation.23 This is a
`rather unique drug–excipient
`interaction in
`which the interaction of the excipient involves
`not only a drug, but also a drug–preservative
`combination that is otherwise incompatible.
`
`Complexing and Dispersing Agents
`
`Cyclodextrins have emerged as very effective ad-
`ditive compounds for solubilizing hydrophobic
`drugs. In the parenteral dosage form area, modi-
`fied cyclodextrins, such as hydroxylpropyl-b-
`cyclodextrin and sulfobutylether-b-cyclodextrin
`have been reported to solubilize and stabilize
`many injectable drugs, including dexamethasone,
`estradiol,
`interleukin-2, and other proteins
`and peptides24 without apparent compatability
`problems.25 There are still only a few approved
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 5 of 18
`
`

`

`2288
`
`AKERS
`
`oleate, isopropyl myrsistate, glycofurol, and fixed
`vegetable oils (e.g., peanut, corn, cottonseed,
`sesame, soybean, castor, arachis). There are
`several oily injectable solutions and suspensions
`used as sustained-release formulations (Clo-
`pixol1, Haldol Decanoate1, Deca-Durabolin1,
`Modecate1, Depixol1, and others).31 Oils are
`generally compatible with lipophilic drugs and
`excipients. However, formulation scientists must
`be aware of the potential for oils to be absorbed by
`rubber closure materials.32,33 Oils can contain
`unacceptable impurities (e.g., gossypol in cotton-
`seed oil, saturated fatty materials, unsaponifi-
`able materials, other organic residuals), but the
`United States Pharmacopeia and other compen-
`dia specify limits on these impurities.
`Highly purified sesame oil was found to
`improve the long-term stability of lidocaine.34
`Lidocaine in sesame oil that was not purified was
`degraded and formed crystals because of oxida-
`tion products such as hydroperoxides and impu-
`rities such as mono- and di-glycerides, free fatty
`acids, plant sterols, and the colorants chlorophyll
`and carotene).
`Soybean oil is the preferred oil in parenteral
`fatty emulsions. Soybean oil emulsions have been
`studied extensively and have been found to be
`incompatible with calcium chloride, calcium glu-
`conate, magnesium chloride, phenytoin sodium,
`tetracycline hydrochloride, and potentially many
`other drug substances and intravenous infusion
`solutions.35
`Petrolatum is a commonly used ointment base
`for topical ophthalmic ointments. There are no
`known incompatibilities with petrolatum.36
`
`Stability Effects
`
`This section highlights both classical and more
`recent publications that report on both positive
`and negative effects of excipients on drug stability
`in parenteral formulations. Examples of some
`esoteric effects (the stabilization of the drug by
`the excipient was not predicted or expected) of
`excipients stabilizing certain drugs are given in
`Table 4.
`
`Table 4. Esoteric Examples of Excipient Stabilizers
`
`Drug or Brand Name
`
`Stabilizing Agent
`
`Dexamethasone acetate
`Cardiotec
`Albumin
`Stelazine/compazine
`
`Creatine or creatinine
`Gamma cyclodextrin
`Sodium caprylate
`Sodium saccharine
`
`Buffers
`
`Buffer components in parenteral formulations
`can cause stability problems. Phosphate buffer,
`particularly the dibasic phosphate anion, serves
`as a nucleophile that can attack electrophilic
`centers like ester or amide carbonyl groups or
`polarized carbon-nitrogen double bonds.37
`Hasegawa et al.38–42 published several articles
`describing the use of pharmaceutical phosphate
`buffer solutions in the presence of calcium and/or
`aluminum by the use of EDTA in pH ranges of 5 to
`9, carboxylic acids such as citric acid and maleic
`acid in acidic to neutral solutions, and pyropho-
`sphate and lysine hydrochloride in alkaline solu-
`tions. Of course, the best approach is to eliminate
`metal contaminants in solutions or additives by
`techniques such as ion exchange, but often this is
`not practical.
`Zhu et al.43 presented a poster describing a
`study in which an unstable drug needed to be
`buffer stabilized at pH 3. Four buffer systems
`(citrate, glycinate, maleate, and tartrate) were
`studied using the same pH; only one buffer
`(glycinate) did not catalyze drug degradation
`(see Fig. 2). The authors did not speculate on
`why glycinate buffer did not catalyze the degra-
`dation of the drug although low concentrations
`(0.1 M) of all buffers would be acceptable. In a
`similar study, Nakamura et al.44 found that
`
`Figure 2. Rate of hydrolysis of GW280430 (0.2 mg/
`mL) as a function of buffer type and concentration at
`608C. (From Zhu, Merserve, & Floyd, Drug Dev Ind
`Pharm, 28, 135–142, ß 2002 Marcel Dekker, Inc.,
`reproduced with permission.)
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 6 of 18
`
`

`

`Table 5. Stability Data for Minodronic Acid in Different Buffer Systems44
`
`EXCIPIENT–DRUG INTERACTIONS
`
`2289
`
`Buffer
`
`Glycine HCl
`
`Citrate
`
`Succinate
`
`Acetate
`
`Tartrate
`
`Lactate
`
`Maleate
`
`pH
`
`3.0
`5.1
`3.0
`5.0
`3.1
`5.0
`3.1
`5.0
`3.0
`5.0
`3.1
`4.4
`3.1
`5.0
`
`Potency (%) After
`4 Weeks at 608C
`
`Number of Particles >2 mm in 100 mM
`Buffer After 4 Weeks at 608C
`
`78.1
`95.5
`99.5
`101.8
`82.4
`94.4
`74.8
`88.5
`93.4
`95.7
`86.0
`93.9
`87.3
`80.1
`
`1000–9999
`> 10,000
`0–99
`0–99
`1000–9999
`> 10,000
`100–999
`1000–9999
`0–99
`0–99
`100–999
`1000–9999
`1000–9999
`> 10,000
`
`citrate and tartrate buffers maintained both
`chemical and physical (fewer particles) stability
`of minodronic acid in a parenteral aqueous solu-
`tion whereas glycine, succinate, acetate, lactate,
`and maleate buffers did not (see Table 5). Li et al.45
`compared the stability of tezacitabine in three
`buffer systems (phosphate, glycine, and TRIS)
`and found phosphate to be superior. Higher con-
`centrations of phosphate also improved drug stabi-
`lity (see Fig. 3a and b).
`
`Tris buffer, when used in a peptide formulation
`and stored at 708C, will degrade to liberate
`formaldehyde.46 Although this was not seen at
`lower temperatures, formulators need to be aware
`of this possible instability when using this com-
`mon biological buffer.
`Tris buffer will form a stable complex with N-
`nitrosourea anticancer agents and retard the de-
`gradation of these agents.47 However, Tris buffer
`will degrade 5-fluorouracil, causing the formation
`
`Figure 3.
`(a) Effect of buffer on stability of FMdC solution at 10 mg/mL. (b) Effect of
`phosphate buffer concentration on stability of FMdC solution at 10 mg/mL. (From Li,
`Chang, Pan, & Jones, AAPS Pharm Sci, (S) 3, ß AAPS Denver, reproduced with
`permission.)
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 7 of 18
`
`

`

`2290
`
`AKERS
`
`of two degradation products that can cause
`serious-to-lethal cardiotoxicities.48
`Sometimes the cationic species in a buffer sys-
`tem matters. Sarciaux et al.49 found that a sodium
`phosphate buffer system consistently resulted in
`more turbid reconstituted solutions of bovine
`immunoglobulin (IgG) than a potassium phos-
`phate buffer system at the same concentration.
`The authors believed that this effect was not
`attributable to a pH shift sometimes seen during
`freezing as a result of crystallization of sodium
`phosphate. This is because sodium chloride-
`containing formulations also showed substan-
`tially higher levels of aggregation compared
`with potassium chloride-containing formulations.
`Bovine IgG will denature at
`the ice/freeze-
`concentrate interface that is irreversible after
`freeze-drying and reconstitution. This ice/freeze-
`concentrate interfacial denaturation is dependent
`on the amount or percentage of protein residing
`at the ice/freeze-concentrate interface and the
`surface area of the freeze-dried solid. Formula-
`tions containing sodium salts showed a higher
`surface area of dried solids than formulations
`containing potassium salts. The higher the surface
`area, the more drug is exposed to the interfacial
`area, resulting in a higher degree of denatura-
`tion and resultant aggregation. When phosphate
`buffers are frozen, selective precipitation of the
`less-soluble buffer component (dibasic sodium
`phosphate) and subsequent pH shift may induce
`protein denaturation.50 In the case of monomeric
`and tetrameric b-galactosidase, both sodium and
`potassium phosphate buffers caused significant
`secondary structural perturbations, greater for
`
`sodium phosphate samples. The addition of suc-
`rose was able to minimize this freeze-dried
`denaturation in phosphate buffers, even if there
`remained large-scale changes in solution pH
`during freezing.
`Histidine was used as a buffer system in an
`experimental formulation containing humanized
`IgG2 monoclonal antibody.51 Histidine under-
`went oxidation and the oxidation products caused
`a significant loss of potency of the monoclonal
`antibody. The antibody also degraded in citrate
`buffer, although not as much as in histidine
`buffer. Histidine buffer oxidizes in the presence
`of peroxides and the source of peroxides in
`these formulations presumably originated from
`Tween 80 also present. Nitrogen overlay inhibited
`the histidine buffer oxidation and enhanced anti-
`body potency.
`
`Antioxidants
`
`Ascorbic acid has been reported to be incompa-
`tible with certain drugs such as penicillin G.52
`However, this is not a direct incompatability of
`two organic molecules, but rather an incompabil-
`ity caused by the pH effects of ascorbic acid,53 as
`specified by Stella.37
`Sodium bisulfite and sodium metabisulfite
`are strongly nucleophilic antioxidants capable of
`catalyzing drug degradation.37 The well-known
`interaction of bisulfite and epinephrine leads to
`degradation of epinephrine. (Scheme 1)
`Epinephrine reacts with bisulfite to form the
`sulfonic acid derivative.54 However, the addition of
`sodium borate complexes the parahydroxybenzyl
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
`Scheme 1.
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 8 of 18
`
`

`

`derivatives and prevents the reaction between
`epinephrine and bisulfite.55 Zinc, copper, and iron
`also will not catalyze any reaction between epine-
`phrine and bisulfite.56 Interestingly, aluminum
`(III) will catalyze epinephrine degradation via a
`complexation effect not seen with these other
`metal ions.56
`Sodium bisulfite also has been reported to
`dehalogenate uracil-type molecules57 and cause
`ester hydrolysis.58 Bisulfite will react with phy-
`sostigmine in aqueous solutions rapidly and
`reversibly, which is dependent both on the total
`bisulfite added and pH.59 Bisulfite will attack
`physostigmine on the optically active carbon-
`10a,60 which is reversed if the mixture is diluted
`and pH adjusted to values greater than pH 6. This
`pH dependency suggested that
`the reaction
`
`2
`reacting with the
`and HSO3
`involved both SO3
`protonated form of physostigmine.
`Sodium metabisulfite inactivates cisplatin in
`solution61 and is incompatible in ophthalmic solu-
`tions containing phenylmercuric acetate, espe-
`cially when autoclaved.62 If dextrose and sodium
`metabisulfite are combined in aqueous solution,
`metabisulfite stability declines.63
`Ascorbic acid is a frequently used antioxidant
`in parenteral formulations. Formulators must be
`aware that ascorbic acid generally is incompatible
`with alkaline solutes, heavy metals, phenylephr-
`ine hydrochloride, pyrilamine maleate, salicyla-
`mide, sodium nitrite, sodium salicylate, and
`theobromine salicylate.64 Kerwin et al.65 found
`that ascorbate ion in sufficient concentrations
`reacted with oxygen producing superoxide that in
`turn caused chemical modification and aggrega-
`tion of recombinant deoxy hemoglobin. Lower
`levels of ascorbate and oxygen and lower solution
`pH combined to eliminate this problem.
`Edetic acid and its salts are used as metal
`chelating agents to aid in stabilization of drugs
`sensitive to metal-catalyzed oxidation and/or
`photolysis. They also can serve to enhance anti-
`microbial activity of formulations. Edetate salts
`are incompatible with zinc insulin, thimersosal,
`amphotericin, and hydralazine hydrochloride.6
`
`Bulking Agents and Lyoprotectants
`
`Several mechanisms have been proposed to ex-
`plain why excipients serve as cryo- or lyo protec-
`tants. The most widely accepted mechanism to
`explain the action of cryoprotection is the pre-
`ferential exclusion mechanism.66 Excipients that
`will stabilize proteins against the effects of freez-
`
`EXCIPIENT–DRUG INTERACTIONS
`
`2291
`
`ing do so by not associating with the surface of
`the protein. Such excipients actually increase the
`surface tension of water and induce preferential
`hydration of the protein. Examples of solutes that
`serve as cryoprotectants by this mechanism in-
`clude amino acids, polyols, sugars, and polyethy-
`lene glycol.
`For lyoprotection, that is, stabilization of pro-
`teins during the drying stages of freeze drying
`and during storage in the dry state, two mechan-
`isms have been generally accepted. One is the
`water-substitute hypothesis67 and the other is
`the vitrification hypothesis.68 Both are legitimate
`theories, but both also have exceptions,
`i.e.,
`neither fully explain the stabilization of proteins
`by excipients during dehydration and dry sto-
`rage.69 The water-substitute hypothesis states
`that a good stabilizer is one that hydrogen bonds
`to the protein just as water would do were it
`present and, therefore, serves as a water sub-
`stitute. Sugars are good water substitutesb which
`is why many freeze-dried protein formulations
`contain sucrose or trehalose.
`The vitrification hypothesis states that excipi-
`ents that remain amorphous (glass formers) form
`a glassy matrix with the protein with the matrix
`serving as a stabilizer. Acceptance in this hypoth-
`esis requires formulators to determine glass
`transition temperatures of formulations to be
`freeze dried and to develop freeze-dry cycles that
`maintain drying temperatures below the glass
`transition temperature. Studies have been pub-
`lished showing that excipient stabilizers that
`crystallized during storage caused degradation
`(typically aggregation and loss of potency) of the
`protein.70–72
`Freeze-dried formulations typically contain
`one or more of the following bulking agents:
`mannitol, lactose, sucrose, trehalose, dextran 40,
`and povidone. These excipients may also serve as
`cryo- and/or lyoprotectants in protein formula-
`tions. Fakes et al.73 studied these bulking agents
`for moisture sorption behavior before and after
`freeze drying. Moisture uptake certainly can affect
`drug stability in the freeze-dried state, particu-
`larly with proteins. They reported the following
`observations for each excipient:
`
`bIt may at first appear contradictory that sugars can serve
`both as cryoprotectants because of being excluded from the
`surface of the protein and as lyoprotectants that hydrogen
`bond to the protein. However, keep in mind that the excluded
`solute concept involves a frozen aqueous system whereas the
`water-substitute concept occurs in a dry system.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 91, NO. 11, NOVEMBER 2002
`
` PFIZER, INC. v. NOVO NORDISK A/S - IPR2020-01252, Ex. 1067, p. 9 of 18
`
`

`

`2292
`
`AKERS
`
`Mannitol
`Crystalline and nonhygroscopic both before
`and after freeze drying
`Total moisture contents of 0.1 to 0.3% w/w
`between 10 and 60% relative humidity (RH)
`Lactose
`Crystalline and nonhygroscopic before lyo-
`philization
`Moisture content 0.86% before lyophilization
`Amorphous after lyophilization with moisture
`content of 1.6%
`Absored moisture rapidly upon exposure to
`high RH
`Converted to crystalline form at 55% RH after
`absorption of 10% moisture
`Desorption of all moisture sorbed by the amor-
`phous form
`Sucrose
`Crystalline and nonhygroscopic before lyophi-
`lization
`Moisture content 0.15% before lyophilization
`Amorphous after lyoph

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