throbber
Pharm Res (2012) 29:3278-3291
`DOI 10.1007/sl 1095-012-0820-7
`
`Effects of Excipients on the Chemical and Physical Stability of Glucagon
`during Freeze-Drying and Storage in Dried Formulations
`
`Wei-Jie Fang• Wei Qi· John Kinzell ·Steven Prestrelski ·John F. Carpenter
`
`Received: 15 December 2011 I Accepted: 22June 2012 /Published online: 6.July 2012
`CCl Springer Science+ Business Media, LLC 2012
`
`ABSTRACT
`Purpose To evaluate the effects of several buffers and exci(cid:173)
`pients on the stability of glucagon during freeze-drying and
`storage as dried powder formulations.
`Methods The chemical and physical stability of glucagon in
`freeze-dried solid formulations was evaluated by a variety of
`techniques including mass spectrometry (MS), reversed phase
`HPLC (RP-HPLC). size exclusion HPLC (SE-HPLC). infrared (IR)
`spectroscopy, differential scanning calorimetry (DSC) and turbidity.
`Results Similar to protein drugs, maintaining the solid amor(cid:173)
`phous phase by incorporating carbohydrates as well as addition
`of surfactant protected lyophilized glucagon from degradation
`during long-term storage. However, different from proteins,
`maintaining/stabilizing the secondary structure of glucagon was
`not a prerequisite for its stability.
`Conclusions The formulation lessons learned from studies of
`freeze-dried formulations of proteins can be applied successfully
`to development of stable formulations of glucagon. However,
`peptides may behave differently than proteins due to their small
`molecule size and less ordered structure.
`
`KEY WORDS excipients · freeze-drying · peptides · solid
`states · stability
`
`W.-J. Fang· W. Qi · j. F. Carpenter(~)
`Department of Pharmaceutical Sciences
`University of Colorado Denver
`Aurora, CO 80045, USA
`e-mail: john.Carpenter@ucdenver.edu
`
`J. Kinzell · S. Prestrelski
`Xeris Pharmaceuticals, Inc.
`San Rafael, CA 9480 I, USA
`
`Present Address:
`W.-J. Fang
`Zhejiang Hisun Pharmaceutical Inc.
`46 Waisha Rd, Jiaojiang District,
`Taizhou, Zhejiang 3 I 8000, China
`
`~Springer
`
`ABBREVIATIONS
`Abbreviations used for amino acids follow the rules of the IUPAC(cid:173)
`IUB Joint Commission of Biochemical Nomenclature (Eur. j. Bio(cid:173)
`chem. 1984 138, 9-37)
`CD
`cyclodextrin
`differential scanning calorimetry
`DSC
`ESl-MS
`electrospray ionization-mass spectrometry
`HES
`hydroxylethyl starch
`IR
`Infrared
`PES
`polyethylstyrene
`PS
`polysorbate
`RP-HPLC
`reversed-phase high-performance liquid
`chromatography
`size exclusion
`glass transition temperature
`retention time
`
`SE
`Tg
`tR
`
`INTRODUCTION
`
`Peptides have become increasingly important as therapeutic
`products. Currently there are more than 60 approved pep(cid:173)
`tide drugs, and an additional 130 peptide candidates are in
`clinical development (1). As is the case with protein thera(cid:173)
`peutics, peptide drugs are susceptible to both chemical and
`physical degradation, and stabilization or these products is
`challenging (2-5). One approach to achieving suflicient
`stability of a biological product is to develop a freeze-dried
`formulation (6, 7). There is an extensive literature describing
`the capacities of various excipients to stabilize proteins dur(cid:173)
`ing the freeze-drying process and long-term storage in dried
`formulations, as well as the mechanisms for such stabiliza(cid:173)
`tion (6--8). For peptide drugs there is a more limited litera(cid:173)
`ture on stabilization in freeze-dried formulations (9-12).
`Therefore, the goal of the current study was to gain forther
`NPS EX. 2049
`CFADv. NPS
`IPR2015-0 I 093
`Page 1
`
`CFAD Exhibit 1046
`CFAD v. NPS
`IPR2015-01093
`
`

`
`Effects of Excipients on Glucagon Stability in Solid
`
`insights into how the lessons learned from protein therapeu(cid:173)
`tics apply to development of stable freeze-dried formulations
`of peptide drugs.
`Typically, an essential stabilizer in a freeze-dried protein
`formulation is a nonreducing disaccharide such as sucrose or
`trehalose (6, 7). These sugars can inhibit protein unfolding
`dming the freezing and drying steps of freeze-drying, as well
`as provide a glassy matrix that is important for long-term
`storage stability of the dried product (6, 7). For drugs that are
`formulated at acid pH, sucrose has the disadvantage of
`being susceptible to acid-catalyzed hydrolysis forming re(cid:173)
`ducing sugars glucose and fructose,( 13) which can chemical(cid:173)
`ly degrade proteins or peptides via the l\!Ialliard reaction
`( 13). For example, the Malliard reaction has been reported
`when glucagon was formulated with the reducing disaccha(cid:173)
`ride lactose ( 14'). Trehalose is more resistant to acidic hy(cid:173)
`drolysis than sucrose and is more suitable for formulations at
`low pH (15,16).
`The inclusion of a polymer such as hydroxylethyl starch
`(HES) to a lyophilized protein formulation may improve
`long-term storage stability of the protein in formulations
`that also contain sucrose or trehalose (17). The increased
`stability is observed because HES can form glassy matrix
`with very high T g (i.e. >200°C). However, HES alone
`usually fails to confer stability to dried proteins because it
`does not inhibit protein unfolding during freeze-drying, and
`the rate of degradation during storage is greatly increased
`for unfolded proteins (6, 7, 1 7).
`The non-ionic surfactants polysorbate 20 and 80 have been
`used extensively as excipients in freeze-dried and aqueous
`solution formulations of proteins due to their ability to reduce
`protein aggregation (18-20). This protective effect ha5 been
`att1ibuted to several different mechanism5 including compet(cid:173)
`ing with protein molecules for interfaces, increasing thermo(cid:173)
`dynamic stability of the native state through binding to the
`protein, fostering refolding and reducing the concentration of
`protein molecules in a stagnant boundary during rehydration
`of dried formulations (20). Cyclodextrins also have been
`shown to reduce protein and peptide aggregation by compet(cid:173)
`ing for the air-water interface (21) and by binding to the
`hydrophobic residues of proteins or peptides such as glucagon
`(14,,22).
`The potential stabilizing effects of each of these classes of
`protein stabilizers for small peptides in freeze-dried formu(cid:173)
`lations are not well understood. Among therapeutic pepti(cid:173)
`des, insulin has been most studied in dried formulations.
`Altl1ough insulin is different from most other peptides in
`that it has relatively well-defined tertiary and quaternary
`structures, it is still instructive to consider results for it in the
`broader context of peptide stabilization. Previous work has
`shown that insulin in freeze-dried formulations exhibited
`many of the same pathways of degradation (i.e. aggregation
`and deamidation) as in aqueous solution (9, 10). In one
`
`,<
`
`3279
`
`study, t11e degradation rate was dependent on the pH of
`aqueous solution p1ior to freeze-drying as well as the water
`content of the freeze-dried cake (9,10). Covalent dimeriza(cid:173)
`tion of human insulin was substantially decreased by incor(cid:173)
`poration into a glassy matrix of trehalose, presumably by
`inhibiting structural perturbation of the peptide, reducing
`molecular mobility in the dried formulation and physically
`separating the insulin molecules ( 10). In another study,
`insulin freeze-dried with trehalose exhibited a substantially
`lower local dynamics (P-relaxation) and lower degradation
`rates (i.e. deamidation and dimerization) than insulin freeze(cid:173)
`dried with dextran ( 12). The authors speculated P-relaxation
`of insulin was reduced because of hydrogen bonding 'vith
`trehalose, and thus chemical degradation was inhibited. On
`the other hand, dextran cannot form hydrogen bonds with
`insulin as readily due to steric hindrance, and therefore
`reduction of P-relaxation was not present in the dextran
`formulation.
`The study of stability in freeze-dried formulation with
`other peptides is rather limited. For example, in one study,
`the level of mannitol freeze-dried formulations of atrial
`natriuretic peptide (ANP) affected the amount of multimers
`formed during storage. The authors speculated that in the
`less stable formulations, which had higher mannitol levels,
`mannitol crystalized and increased ilie water moisture con(cid:173)
`tent in the amorphous phase ( 11 ).
`Glucagon (Fig. 1 ), the focus of the current study, is a
`polypeptide hormone composed of 29 amino acid residues
`that is currently used for the emergency treatment ofinsulin(cid:173)
`induced hypoglycemia (23). Glucagon is known for its pro(cid:173)
`pensity to degrade both chemically (i.e., hydrolysis and
`oxidation)(24-26) and physically (i.e., aggregation)(27 ,28);
`including formation of aggregates during freeze-drying and
`rehydration in the absence of stabilizing excipients (29).
`Therefore, it is an excellent model peptide to evaluate the
`effects of different buffers, the surfactant polysorbate 20,
`trehalose, HES and P-CD on stability during freeze-drying
`and storage in dried formulations.
`
`MATERIALS AND METHODS
`
`Materials
`
`All chemicals were of reagent grade or higher quality. Glu(cid:173)
`cagon was purchased from the American Peptide Company
`(Sunnyvale, CA). P-CD was purchased from Cyclodext1in
`Technologies Development, Inc. (High Springs, FL). Tre(cid:173)
`halose dihydrate was purchased from J. T.Baker (Philips(cid:173)
`burg, NJ). HES (Viastarch, MW 200KDa) was purchased
`from Fresenius (Graz, Austria). Phosphoric acid, citric acid,
`glycine, 2M hydrogen chloride, sodium chloride, sodium
`hydroxide, acetonitrile, hydrogen peroxide, potassium
`
`~Springer
`
`Page2
`
`

`
`3280
`
`Fang et al.
`
`His-Ser-Gln-Gly-Thr-Phe-Thr-Ser-Asp9-Tyr-Ser-Lys-Tyr-Leu-Asp 15 -Ser-Arg-Arg-Ala-Gln-
`
`Fig. I Primary structure of glucagon. The three Asp and Met residues are shown in bold.
`
`Asp21 -Phe-Val-Gln-Trp-Leu-Met27-Asn-Thr
`
`bromide and polysorbate 20 were purchased from Fisher
`Scientific (Hampton, NH). Nylon membrane filters
`(0.4·5 µm) were purchased from Whatman (Maidstone, Eng(cid:173)
`land). Polyethylstyrene (PES) membrane filters (0.2 µm)
`were purchased from Millipore (Billerica, MA). 3-ml lyoph(cid:173)
`ilization vials (borosilicate glasses, type 8412-B) and gray
`butyl stoppers (Product# 10123524) were purchased from
`West Pharmaceutical (Lionville, PA). Other reagents and
`chemicals were purchased from Sigma-Aldrich (Jvlilwaukee,
`WI).
`
`Acidic Degradation of Glucagon in Aqueous Solution
`
`Glucagon (0.5 mg/ml) was dissolved in 5 mM sodium phos(cid:173)
`phate bufler (pH=3.0) and incubated for 5 days at 60°C.
`Aliquots (0.5 mL) of the solution were removed at various
`times and centrifoged at 14,500 rpm for 10 min. The
`supernatant was analyzed by reversed-phase high(cid:173)
`performance liquid chromatography (RP-HPLC) as de(cid:173)
`scribed below. Samples were also analyzed by electrospray
`ionization-mass spectrometry (ESI-MS; see below).
`
`Hydrogen Peroxide-Induced Oxidation of Glucagon
`in Aqueous Solution
`
`We followed the USP method (USP 30 Official Monograph
`for Glucagon, 2007) for forced oxidation of glucagon. Glu(cid:173)
`cagon (1 mg/ml) was oxidized by incubation in 0.6% hy(cid:173)
`drogen peroxide (H20 2), 20% acetonitrile and 80% water
`with 10 mM HCl at 4°C for 210 min. Aliquots were re(cid:173)
`moved at various times and analyzed by RP-HPLC using
`the method described below. The oxidation products were
`also analyzed by ESI-MS as well as tandem MS (MS-MS;
`see below).
`
`ESl-MS and MS-MS Analysis of Glucagon
`and its Degradation Products
`
`temperature. For MS-MS experiment~ on glucagon and
`oxidized glucagon, various ml z components were selected
`and fragmented with suitable collision energy to have rea(cid:173)
`sonable amount of peaks following fragmentation.
`
`Sample Preparation for Freeze-Drying
`
`Glucagon (10 mg/mL) was dissolved in three different buf(cid:173)
`fers (glycine hydrochloride, sodium phosphate, and sodium
`citrate buflers, 5 mM, pH 3.0). The solution was then mixed
`in a l: l (v /v) ratio with various excipient solutions (prepared
`at twice the desired concentration using corresponding bufl~
`er) to obtain a final glucagon concentration of 5 mg/mL and
`the final desired excipient concentration of 0.0 l % for poly(cid:173)
`sorbate 20 and 10 mg/mL for the carbohydrates. The
`solution was then filtered through 0.2 µm Millipore PES
`membrane. Sample preparation was conducted in a 4°C
`cold room. The glucagon concentration and the purity were
`determined by RP-HPLC (see below).
`
`Freeze-Drying
`
`The formulations were pipetted (0.3 mL) into 3-ml lyophiliza(cid:173)
`tion vials (13-mm ID) and freeze-dried in a FTS Durastop
`freeze-drier (Stoneridge, NY). For freezing, samples were
`cooled to -40°C at 2.5°C/min and maintained at this tem(cid:173)
`perature for 2 h. Then the shelf temperature was increa~ed to -
`5°C at 2°C/min and held for 2 has an annealing step (29). The
`temperature was then decreased to -30°C at l.5°C/min and
`the chamber pressure was reduced to 8 Pascal. These condi(cid:173)
`tions were maintained during 24 h for primary drying. Then
`the shelf temperature was increased to 40°C at 0.5°C/min and
`held at 40°C for l 0 h for secondary drying. Then, the vials
`were stoppered under vacuum using gray butyl stoppers. None
`of the formulations showed any visual evidence of cake collapse
`following freeze-drying.
`
`Storage Studies
`
`Glucagon and its degradation product~ were analyzed with
`an electrospray-triple quadrupole-time-of~flight mass spec(cid:173)
`trometer (ESI-qTOF-MS) from Applied Biosystems (PE
`SCIEX/ ABI API QSTAR Pulsar i Hybrid LC/MS/
`MSESL). Mass Spectra were acquired by scanning a mass(cid:173)
`to-charge ratios (ml z) range from l 00 to 2000. Eluates
`(l mg/ml glucagon, 50 µl) were injected into the mass
`spectrometer at a flow rate of 5 µI/min. Spray voltage wa~
`set at 4500 V, and capillary temperature was set at ambient
`
`Following freeze-drying, the sample vials were incubated at
`60°C and analyzed after 2 weeks. Triplicate sample vials for
`each formulation were incubated. Control sample vials
`(equivalent to time zero of incubation) were stored at -80°
`C until analysis. Following storage, formulations were first
`rehydrated to 5 mg/mL with water and then diluted to
`l mg/mL glucagon with the corresponding buffer. An ali(cid:173)
`quot of the rehydrated sample (200 µL) was analyzed for
`
`~Springer
`
`Page 3
`
`

`
`Effects of Excipients on Glucagon Stability in Solid
`
`3281
`
`turbidity (see below). Then, the solution was centrifoged
`(14·,500 111mx 10 min) to removed insoluble material. The
`supernatant was diluted 1: 1 with the corresponding buffer
`for RP-HPLC and size exclusion (SE)-HPLC analysis (see
`below).
`
`Differential Scanning Calorimetry (DSC)(JO)
`
`The gJa5s transition temperature (Tg) of dried formulations
`was determined using a Perkin-Elmer Pyris-1 DSC (Nor(cid:173)
`walk, CT). Prior to sample analysis, the instrument was
`calibrated for melting temperature and heat of fusion with
`indium (onset of melting: 156.6°C; heat of fusion: 28.4-5 JI
`g). In a d1y box, the sample vials were opened and about
`1 mg samples of dry powder were placed in hermetically
`sealed aluminum pans. DSC thermograms were collected
`from 25°C up to 250°C (depending on the compositions of
`formulations) at a heating rate of 100°C/min. Baselines
`were determined using an empty pan, and all thermograms
`were baseline corrected. For measurement of T g> the sam(cid:173)
`ples were first heated to above T g to remove thermal histo1y,
`and then cooled back to 25°C and rescanned at 100°C/min.
`The thermogram obtained during the second scan was used
`to measure T g which was determined as the midpoint of the
`transition.
`
`Infrared (IR) Spectroscopy
`
`Infrared spectra of glucagon in aqueous solution and in
`freeze-dried formulations were collected at room tempera(cid:173)
`ture using a Bomem Prata spectrometer (Quebec, Canada)
`purged with d1y nitrogen (31,32). Spectra of glucagon in
`aqueous solution were obtained at a peptide concentration
`of 20 mg/ml (5 mM sodium phosphate buffer, pH 3.0) in a
`6 µm pathlength cell with CaF2 windows. For each sample,
`a 32-scan interferogram was collected in the single-beam
`mode with a resolution of 4 cm - t. Peptide spectra were
`processed to substract absorbance from water vapor and
`the buffer spectrum as previously described (31,32). For IR
`spectra of freeze-dried samples, a mass of dried formulation
`containing about 1 mg of glucagon was mixed with 500 mg
`potassium bromide powder. The mixture was ground gently
`and then annealed into a pellet using a hydraulic press (30).
`The spectra were transformed to second derivatives using
`Bomem Grams/32 software. The final protein spectra were
`smoothed with a seven-point Savitsky-Golay function. For
`comparison of spectra they were area normalized in the
`amide I region ( 1600-1700 cm -l) and overlaid (30).
`
`using a Molecular Devices microplate reader (Sunnyvale,
`CA). The freeze-dried glucagon formulations were first
`rehydrated with water to 5 mg/mL glucagon and then
`diluted with corresponding buffer to 1 mg/mL peptide
`concentration. Prior to analysis, the plates were shaken for
`10 s to mix the solutions in the wells.
`
`Reversed Phase-High Performance Liquid
`Chromatography (RP-HPLC)
`
`Chemical degradation of glucagon formulations was quan(cid:173)
`tified by RP-HPLC, using an Agilent 1100 HPLC system
`(Agilent Technologies, Inc., vValdbronn, Karlsruhe, Ger(cid:173)
`many) with a Thermo Biobasic CS column (5 µm, 250 x
`4.0 mm ID, Waltham, MA) and a Restek VWD Gl3J4.A
`detector (Bellefonte, PA). The mobile phase was 73%
`phosphate-cysteine bufler (pH 2.6) with 27% acetonitrile
`(USP 30 Official Monograph for Glucagon, pp 2230-
`2231, 2007). Before use, mobile phase was filtered with
`0.4·5 µm Nylon membrane filters and degassed. The flow
`rate for the analysis was 1 ml/min, and elution was moni(cid:173)
`tored at 214 nm. The temperature of the column was
`maintained at 37°C. The peak areas of glucagon and its
`chemical degradation products were used to determine the
`chemical degradation of glucagon occurring during freeze(cid:173)
`drying and storage.
`
`Size Exclusion-HPLC (SE-HPLC)
`
`Aggregation of glucagon in different formulations was de(cid:173)
`termined with SE-HPLC, using an Agilent 1100 HPLC
`equipped with a TSK G2000SWx1 gel filtration column
`(5 µm, 300X 7.8 mm ID) and a Restek DAD Gl315A
`detector. The mobile phase was 3.2 mM HCl, 100 mM
`sodium chloride, pH 2.5. Before use, the mobile phase was
`filtered and degassed. The flow rate for the analysis was
`1 ml/min and elution was monitored at 280 nm. The
`percent of monomer remaining in formulations was calcu(cid:173)
`lated based on the monomer area of the formulation sam(cid:173)
`ples compared to that for liquid control sample. Soluble
`aggregates were at minimal levels (<0.5%) in all of the
`samples (data not shown).
`
`RESULTS AND DISCUSSION
`
`Accelerated Acid Degradation of Glucagon
`in Aqueous Solution
`
`Optical Density at 405 nm (OD40s)
`
`The presence of aggregates in rehydrated formulations
`(1 mg/mL glucagon) was assessed by measuring the OD405
`
`Glucagon has an isoelectric point of about 7 and has a high
`solubility at pH values less than 3 or greater than 9. The
`recommended pH range for solutions of the peptide is
`between 2.5 and 3.0 (US pharmacopeia 24, 1999).
`
`~Springer
`
`Page4
`
`

`
`3282
`
`Fang et al.
`
`However, as in the case with many peptide and protein
`drugs, glucagon is susceptible to acid-catalyzed degradations
`(i.e. Asp cleavage) (24-26). To develop an assay to charac(cid:173)
`terize acid-catalyzed hydrolysis of glucagon, the peptide was
`first incubated in 5 mM sodium phosphate buffer (pH=3.0)
`at 60°C and characterized as a fonction of incubation time.
`The commercial glucagon we used had some impurities
`prior to incubation (about 8-9%, see Fig. 3a and its en(cid:173)
`larged version Fig. 3b). During incubation, glucagon under(cid:173)
`went significant further chemical degradation, with 50%
`loss of the parent peptide after incubation for 5 days
`(Figs. 2 and 3d). As expected in the acidic conditions,
`the main degradation products were due to Asp cleavage,
`as confirmed by ESI-MS analysis (Table I). Glucagon has
`three Asp residues and, therefore, six main fragments were
`observed in ESI-MS.
`In RP-HPLC (Fig. 3d), four main peaks (retention time
`(tR) around 7, 14, 19 and 29 min, respectively) and several
`smaller peaks (i.e. lR around 33, 37, 40, and 42 min) were
`observed. The main peaks were probably due to Asp cleav(cid:173)
`age whereas the smaller peaks could be due to Asp cleavage
`and/or deamidation (25,26). Glucagon has three Gln and
`one Asn residue. Deamidated residues were not specifically
`identified in the current study.
`
`Accelerated Oxidation of Glucagon in Aqueous
`Solution
`
`Although oxidation of Met does not impact the binding
`aflinity of glucagon to its receptor (33), the potency the
`oxidized peptide in stimulating glucose production in isolat(cid:173)
`ed rat adipocytes and hepatocytes is decreased (34-). There(cid:173)
`fore, oxidation of the peptide should be monitored and
`minimized. To develop an assay for oxidized glucagon, the
`peptide was incubated in 0.6% (v/v) hydrogen peroxide at
`room temperature (Fig. 4). Samples were removed as a
`function of time and analyzed by RP-HPLC. Two new
`
`'[ 2.0
`
`Cl ns u a 1.9
`Cl c ·c: 1.8
`"itj
`~
`0:: 1.7 c
`.3
`
`Cl
`
`0
`
`4
`2
`Time (days)
`
`6
`
`Fig. 2 Degradation of glucagon in 5 mM sodium phosphate buffer pH 3.0 at
`60°C. The % remaining glucagon was determined from chromatograms
`obtained by RP-HPLC. Error bars indicating SD are smaller than the symbols.
`
`~Springer
`
`peaks with similar tR and almost identical areas were ob(cid:173)
`served (tR around 8-9 min, Fig. 3c). Almost all of the parent
`peptide had decomposed after 210 min of incubation
`(Fig. 4). The oxidation products had molecular weights of
`[Glucagon+ 16] (calculated mass=3498.6; observed: [M +
`2 H] 2+=1750.2, [M+3 H] 3+=1167.l, [Iv1+4 H] 4+=875.6,
`[M +5 HJ 5+=700.7), suggesting one residue of glucagon was
`oxidized. MS/MS analysis (Table II) of the oxidation prod(cid:173)
`ucts showed the Met residue at position 27 was oxidized.
`The two peaks in the RP-HPLC chromatograms were prob(cid:173)
`ably due to two diastereomers formed with oxidation of the
`Met residue. Even though glucagon also has several other
`amino acids susceptible to oxidation (e.g. His, Trp, Tyr, and
`Phe), no degradation of these residues by hydrogen peroxide
`was observed.
`
`Effects of Buffers and Excipients on Glucagon's
`Stability in Freeze-Dried Formulations
`
`To determine formulation effects on stability of freeze-dried
`glucagon formulations, we evaluated effects of three hullers
`(glycine hydrochloride, sodium phosphate and sodium cit(cid:173)
`rate bullers, 5 mM, pH 3.0), polysorbate 20 and three
`carbohydrate excipients, trehalose, HES and P-CD. An
`accelerated degradation condition (storage at 60°C for
`2 weeks, followed by rehydration) was used to evaluate the
`stability of the peptide in the freeze-dried formulations.
`Important physical properties (peptide secondary structure
`and T g) were also characterized for each freeze-dried
`formulation.
`The T g of the freeze-dried formulations was determined
`by DSC (Table III). When there was no excipient, the Tg of
`dry powders was between l 22°C and l 4·0°C, depending on
`the buffer used in formulation (Table III). A~ expected, the
`presence ofpolysorbate 20 (0.01%w/v) did not have much
`effect on Tw
`For formulations with glass forming carbohydrates, the
`Tg was highest for HES and P-CD, followed by the mixture
`of trehalose and HES, and then trehalose alone (Table III).
`These values were consistent with the T g of the
`corresponding pure carbohydrates and their mass ratios in
`the formulations. The huller also had a substantial ellect on
`T g' especially in the cases where HES and P-CD were used
`as excipients (Table III). The cause(s) of the huller effect was
`not investigated forther.
`Infrared spectroscopic analysis of glucagon formulations
`was used to evaluate the ellects of freeze-drying and storage
`on glucagon's secondary structure, by comparing spectra for
`the peptide in the dried formulations to that for the native
`peptide in aqueous solution. The main secondary structure
`of native glucagon is a mixture of a-helix and random coil
`(35,36). Correspondingly, the infrared spectrum for native
`glucagon in aqueous solution has main absorbance around
`
`Page 5
`
`

`
`Effects of Excipients on Glucagon Stability in Solid
`
`Fig. 3 RP-HPLC
`chromatograms of glucagon and
`its chemical degradation
`products. (a) Control sample; (b)
`expanded scale of a; (c) oxidized
`glucagon by hydrogen peroxide;
`(d) degraded in 5 mM sodium
`sodium phosphate buffer, pH 3.0,
`at 60°C for 5 days; (e) following
`incubation of dry powder (freeze(cid:173)
`dried in sodium phosphate buffer)
`at 60°C for 2 weeks (scale was
`expanded for improved visual
`resolution of peaks).
`
`GOO
`
`a
`
`glucagon
`/
`
`I
`\ l
`
`5
`
`10 15 20 25 30 35 40 45
`Retention tirre (rrin)
`
`50
`
`20
`
`e 15
`
`<ol'
`
`l\i r 0
`
`5
`
`10
`
`b
`
`3283
`
`glucagon
`
`/
`
`JV\______
`
`15 20 25 30 35 40 45 50
`Retention tirre (rrin)
`
`d
`
`glucagon
`/
`
`c
`
`/
`
`oxidiz.ed glucagon
`
`2SO
`
`e 200
`
`<ol' 1: 0
`~: e
`/IJ~
`f:uV •lAlL
`
`5
`
`10 15 20 25 30 35 40 45
`Retention tirre (rrin)
`
`50
`
`5
`
`10 15 20 25 30 35 40 45 50
`Retention tirre (rrin)
`
`5
`
`10 15 20 25 30 35 40 45 50
`Retention tirre (rrin)
`
`1657 cm- 1 (Fig. 5). The peak is broad, presumably due to
`high flexibility of backbone conformation.
`
`For comparison of the native peptide spectrum to that of
`folly denatured and aggregated glucagon, the peptide
`
`Table I ESl-MS Analysis of Glucagon Hydrolysis Products After Incubating at 5 mM Sodium Phosphate Buffer, pH 3.0 at 60°C for 2 days
`
`Degradation product
`
`Glucagon [ 1-9]
`His-Ser-Gln-Gly-Thr-Phe-Thr-Ser-Asp
`Glucagon [I 0-29]
`Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Arg-Arg-Ala-Gln-Asp-Phe-Val-Gln-Trp-Leu-Met-Asn-Thr
`
`Glucagon [ 1-1 5]
`His-Ser-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp
`
`Glucagon [ 16-29]
`Ser-Arg-Arg-Ala-Gln-Asp-Phe-Val-Gln-T rp-Leu-Met-Asn-Thr
`
`Glucagon [1-21]
`His-Ser-Gln-Gly-Thr-Phe-Thr-Ser-Asp-Tyr-Ser-Lys-Tyr-Leu-Asp-Ser-Arg-Arg-Ala-Gln-Asp
`
`Glucagon [22-29]
`Phe-Val-Gln-Trp-Leu-Met-Asn-Thr
`
`ESl-MS (m/z)
`
`Calculated
`
`[M +Ht =978.4
`[M + 22+=490.2
`[M + 2 Hf+= 1261 .6
`[M+3 H]3+=841.4
`[M+4 H]4+=631.3
`[M+2 H]2+=874.9
`[M+3 H]3+=583.6
`[M+2 H]2+=876.4
`[M+3 Hf'=584.6
`[M+3 H]3+=821.7
`[M + 4H]4+ =616.5
`[M +Ht= 1038.5
`[M + 2H]2+=5 I 9.8
`
`Observed
`
`[M + Ht=978.4
`[M + 2 H]2+ =490.2
`[M+2 Hf'=1261.6
`[M + 3 H]3+=84 I .4
`[M +4 H]4+=63 I .3
`[M + 2 H]2+=874.9
`[M+3 H]3+=583.6
`[M+2 H]2+=876.4
`[M+3 H]3'=584.6
`[M+3 H]3+=821.7
`[M + 4H]4+ =616.5
`[M+Ht= 1038.5
`[M + 2Hf+=5 I 9.8
`
`if:! Springer
`
`Page6
`
`

`
`3284
`
`g2.o
`Cl
`Ill u
`::I
`(5
`gi 1.5
`·2
`'iii
`E
`Cl)
`0::
`~ 1.0
`Cl
`0
`....I
`
`0
`
`50
`
`150
`100
`Time (min)
`
`200
`
`250
`
`Fig. 4 Oxidative degradation of glucagon with 0.6% H102 at 4°C. The
`% remaining glucagon was determined from chromatograms obtained
`by RP-HPLC.
`
`solution was incubated in a boiling water bath for 20 min and
`then freeze-dried. It ha~ been reported that upon fibrillation,
`the main secondary structure of glucagon is converted into
`intermolecular P-sheet (3 7 ,38). In the IR spectrum of boiled
`and freeze-dried glucagon the peak intensity around
`165 7 cm - I was greatly decreased and a new peak around
`1630 cm- 1 was observed (Fig. 5), suggesting intermolecular P(cid:173)
`sheet was formed between glucagon molecules during
`heating-induced aggregation.
`For analysis of the freeze-dried formulations, first the
`infrared spectra of glucagon formulations freeze-dried in
`glycine hydrochloride buffer were studied (Figs. Ga and
`7a). The dominant spectral feature affected by formulation
`was the intensity or the band at l G5 7 cm - I. Therefore this
`parameter was used to compare results for the difforent
`formulations (Fig. 7). Compared to the spectrum of the
`native, aqueous peptide, the band intensity was only slightly
`decreased in the spectrum for the peptide freeze-dried in
`bufler alone. There was also no increase o!' absorbance
`around 1630 cm- 1-a strong band in the spectrum of
`denatured/aggregated glucagon-suggesting no significant
`amount of intermolecular P-sheet was formed during freeze(cid:173)
`drying of glucagon in glycine buffer.
`The presence of polysorbate 20, HES or P-CD alone in
`the formulation did not result in substantial difference in the
`
`Fang et al.
`
`peak intensity at 1657 cm-1 compared to that for the peptide
`freeze-dried in buffer alone (Figs. Ga and 7a). In contrast,
`freeze-drying in formulations containing trehalose resulted
`in a substantial increase in band intensity due to narrowing
`of the band. This result is presumably because of an increase
`in the structural compaction of the peptide and a more
`narrow distribution of conformational substates than that
`present in aqueous solution. The mixture of trehalose and
`HES also preserved tl1e a-helix structure of glucagon, but
`was less effective compared with the formulation in which
`trehalose was the only excipient.
`Trehalose eflectively preserved the secondary structure of
`glucagon probably by hydrogen bonding to the peptide in
`place of water during drying, as has been observed with
`many proteins (G, 7). HES or P-CD probably could not form
`effective hydrogen-bonds with dried glucagon and, there(cid:173)
`fore, did not have a protective effect on the secondary
`structure of glucagon.
`The effects of incubation (G0°C for 2 weeks) of the freeze(cid:173)
`dried formulations prepared in glycine bufler on glucagon's
`secondary structure were also evaluated (Figs. Gb and 7b).
`Interestingly, the a-helix peak intensity for most formula(cid:173)
`tions was increased following incubation, suggesting that the
`secondary structure of glucagon was more ordered. The
`cause of this effect is not known.
`The eflects of excipients in sodium phosphate buffer were
`similar to those seen in glycine hydrochloride buffer (Fig. 7),
`with the greatest intensity of the l G5 7 cm - I band noted in
`freeze-dried formulations containing trehalose. Also, the
`peak intensities in the glucagon spectra were increased by
`incubation of the freeze-dried formulations at 60°C for
`2 week~.
`With sodium citrate buffer the eflects of excipients also
`were similar to those seen in glycine bu!Ier (Fig. 7a), with the
`greatest intensity of the 165 7 cm - I band noted in freeze(cid:173)
`dried formulations containing trehalose (Fig. 7a). However,
`unlike the results noted in formulations in glycine and sodi(cid:173)
`um phosphate buffers in which the presence of P-CD did not
`change the secondary structure compared to the formula(cid:173)
`tions without any excipient, there was increased band
`
`Table II MS-MS Analysis of Glu(cid:173)
`cagon and Oxidized Glucagon
`
`Parent species charge0
`
`Glucagonb
`
`Oxidized glucagonb
`
`5
`
`4
`
`3
`2
`
`3 Parent species with 2-5 charges
`were chosen for fragmentation.
`bThe main fragments were shown
`with charge state. The data sug(cid:173)
`gested the oxidation is in Met27
`residue
`
`~Springer
`
`365.2 (+I): y3 ion Met-Asn-Thr
`246.0 ( + I): Met-Asn
`780.4 (+4): b ion ofGlu(l-26)
`813.1 ( +4): b ion of Glu(l-27)
`365.2 ( + I): y3 ion Met-Asn-Thr
`246.0 ( + I): Met-Asn
`84 i .9 ( +4): b ion of Glu(l-28)
`876.5 ( +2): y ion of Glu(l6-29)
`1261. I (+2): y ion of Glu(I0-29)
`
`381.1 (+I) y3 ion of Met(O)-Asn-Thr
`262. I ( + I): Met(O)-Asn
`780.4 (+4): b ion ofGlu(l-26)
`817.2 ( +4): b ion of [O]Glu(l-27)
`381.1 (+I): y3 ion of Met(O)-Asn-Thr
`262. I ( + I): Met(O)-Asn
`845.9 ( +4): b ion of [O]Glu(l-28)
`884. 4 ( + 2): y ion of [O]Glu( 16-29)
`1269.6 ( + 2): y ion of [O]Glu(I 0-29)
`
`Page 7
`
`

`
`Effects of Excipients on Glucagon Stability in Solid
`
`3285
`
`Table Ill Composition and Tg (0C) of Freeze-Dried Glucagon Formulations
`
`Excipient
`
`Buffer0
`
`None
`
`PS 20
`
`Trehalose
`
`HES
`
`Tre-HES
`
`13-CD
`
`Glycine
`
`Glucagon: 90%
`Glycine: I 0%
`Glucagon: 89%
`Glycine: 9%
`PS 20: 2%
`Glucagon: 32%
`Glycine: 4%
`Tre: 64%
`Glucagon: 32%
`Glycine: 4%
`
`HES: 64%
`Glucagon: 32%
`Glycine: 4%
`Tre: 32%
`HES: 32%
`Glucagon: 32%
`Glycine: 4%
`
`13-CD: 64%
`
`133.1 ± 1.2 (3)
`
`140.0(1)
`
`105.3 ±0.6
`
`207.7± 1.8
`
`124.1 ±6.4
`
`218.2± 11.3
`
`Phosphate
`
`Glucagon: 90%
`Phosphate: I 0%
`Glucagon: 89%
`Phosphate: 9%
`PS 20: 2%
`Glucagon: 32%
`Phosphate: 4%
`Tre: 64%
`Glucagon: 32%
`Phosphate: 4%
`HES: 64%
`Glucagon: 32%
`Phosphate: 4%
`Tre: 32%
`HES: 32%
`Glucagon: 32%
`Phosphate: 4%
`
`13-CD: 64%
`
`140.1 ±0.7
`
`134.8±0.8
`
`11

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