throbber
Investigation of Histidine Stabilizing Effects on LDH During
`Freeze-Drying
`
`ANAS AL-HUSSEIN,1 HENNING GIESELER1,2
`
`1Division of Pharmaceutics, Freeze Drying Focus Group, University of Erlangen, Erlangen 91058, Germany
`
`2GILYOS GmbH, Wuerzburg 97076, Germany
`
`Received 2 September 2012; revised 29 November 2012; accepted 30 November 2012
`
`Published online 20 December 2012 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23427
`
`ABSTRACT: The objective of this study was to investigate the effect of histidine on the stability
`of the model protein lactate dehydrogenase (LDH) during freeze-drying. Several parameters
`were varied including pH of the bulk solution, histidine concentration, and performance of an
`annealing step during freezing. First, histidine was used as a buffer in the protein formulations
`and compared with “conventional” potassium phosphate and citrate buffer systems. For this
`purpose, sucrose or mannitol was used as stabilizers. Second, the possibility of using histidine
`as both buffer and stabilizer (cryoprotectant and lyoprotectant) in the protein formulations was
`evaluated with focus on protein stability and the physical state of histidine in the final product,
`in addition to cake elegance. Protein stability was evaluated both functionally by measuring the
`activity recovery of the model protein LDH after freeze-drying and structurally by analyzing the
`protein secondary structure. LDH showed improved stability in histidine buffer in comparison
`with other buffers. Protein stability and the tendency of histidine to crystallize during freeze-
`drying were pH dependent. Annealing destabilized LDH and resulted in a decrease of the
`activity recovery. However, the extent of protein destabilization caused by annealing appears to
`be also pH dependent. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Association
`J Pharm Sci 102:813–826, 2013
`Keywords:
`freeze drying/lyophilization; stability; proteins; excipients; physical characteriza-
`tion; buffers
`
`INTRODUCTION
`Advances in biotechnology over the last decades re-
`sulted in an increasing number of therapeutic pro-
`teins. Protein-based products are likely to represent
`four of the five top-selling drugs globally by 2013.1 The
`maintenance of protein stability and efficacy in the
`dosage form presents a great challenge to the phar-
`maceutical industry. Because of their limited stability
`in an aqueous environment, proteins often need to be
`converted into solid state to achieve an acceptable
`shelf life as pharmaceutical products.2 The most com-
`monly used method for manufacture of solid protein
`pharmaceuticals is freeze-drying (lyophilization).3,4
`However, the freeze-drying process generates many
`stresses during both freezing and drying, which may
`cause the loss of protein bioactivity. Therefore, a range
`
`Correspondence to: Henning Gieseler (Telephone: +49-931-
`90705678; Fax: +49-931-90705679; E-mail: info@gilyos.com)
`Journal of Pharmaceutical Sciences, Vol. 102, 813–826 (2013)
`© 2012 Wiley Periodicals, Inc. and the American Pharmacists Association
`
`of excipients can be added to the protein formulation
`to overcome these stresses and to improve protein sta-
`bility during freeze-drying and storage. This goal is
`commonly achieved by using amorphous excipients
`to serve as protein stabilizer during both freezing
`(cryoprotectant) and drying (lyoprotectant).3,4 One of
`the most widely accepted protein stabilization mecha-
`nisms during freezing is preferential exclusion, which
`means that the excipient is preferentially excluded
`from the surface of the protein. Thereby the free
`energy required for denaturation is increased and
`the native structure of the protein is stabilized.5
`During the drying phase, the protein is stabilized
`by the water replacement mechanism and by the
`formation of a viscous glassy state. The water re-
`placement mechanism involves the formation of hy-
`drogen bonds between a protein and an excipient
`to satisfy the hydrogen-bonding requirement of po-
`lar groups on the protein surface.6 The formation
`of an amorphous viscous glass during freeze-drying
`and the corresponding extremely high viscosity
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`813
`
`Page 1
`
`NPS EX. 2060
`CFAD v. NPS
`IPR2015-01093
`
`

`
`Investigation of Histidine Stabilizing Effects on LDH During
`Freeze-Drying
`
`ANAS AL-HUSSEIN,1 HENNING GIESELER1,2
`
`1Division of Pharmaceutics, Freeze Drying Focus Group, University of Erlangen, Erlangen 91058, Germany
`
`2GILYOS GmbH, Wuerzburg 97076, Germany
`
`Received 2 September 2012; revised 29 November 2012; accepted 30 November 2012
`
`Published online 20 December 2012 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23427
`
`ABSTRACT: The objective of this study was to investigate the effect of histidine on the stability
`of the model protein lactate dehydrogenase (LDH) during freeze-drying. Several parameters
`were varied including pH of the bulk solution, histidine concentration, and performance of an
`annealing step during freezing. First, histidine was used as a buffer in the protein formulations
`and compared with “conventional” potassium phosphate and citrate buffer systems. For this
`purpose, sucrose or mannitol was used as stabilizers. Second, the possibility of using histidine
`as both buffer and stabilizer (cryoprotectant and lyoprotectant) in the protein formulations was
`evaluated with focus on protein stability and the physical state of histidine in the final product,
`in addition to cake elegance. Protein stability was evaluated both functionally by measuring the
`activity recovery of the model protein LDH after freeze-drying and structurally by analyzing the
`protein secondary structure. LDH showed improved stability in histidine buffer in comparison
`with other buffers. Protein stability and the tendency of histidine to crystallize during freeze-
`drying were pH dependent. Annealing destabilized LDH and resulted in a decrease of the
`activity recovery. However, the extent of protein destabilization caused by annealing appears to
`be also pH dependent. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Association
`J Pharm Sci 102:813–826, 2013
`Keywords:
`freeze drying/lyophilization; stability; proteins; excipients; physical characteriza-
`tion; buffers
`
`INTRODUCTION
`Advances in biotechnology over the last decades re-
`sulted in an increasing number of therapeutic pro-
`teins. Protein-based products are likely to represent
`four of the five top-selling drugs globally by 2013.1 The
`maintenance of protein stability and efficacy in the
`dosage form presents a great challenge to the phar-
`maceutical industry. Because of their limited stability
`in an aqueous environment, proteins often need to be
`converted into solid state to achieve an acceptable
`shelf life as pharmaceutical products.2 The most com-
`monly used method for manufacture of solid protein
`pharmaceuticals is freeze-drying (lyophilization).3,4
`However, the freeze-drying process generates many
`stresses during both freezing and drying, which may
`cause the loss of protein bioactivity. Therefore, a range
`
`Correspondence to: Henning Gieseler (Telephone: +49-931-
`90705678; Fax: +49-931-90705679; E-mail: info@gilyos.com)
`Journal of Pharmaceutical Sciences, Vol. 102, 813–826 (2013)
`© 2012 Wiley Periodicals, Inc. and the American Pharmacists Association
`
`of excipients can be added to the protein formulation
`to overcome these stresses and to improve protein sta-
`bility during freeze-drying and storage. This goal is
`commonly achieved by using amorphous excipients
`to serve as protein stabilizer during both freezing
`(cryoprotectant) and drying (lyoprotectant).3,4 One of
`the most widely accepted protein stabilization mecha-
`nisms during freezing is preferential exclusion, which
`means that the excipient is preferentially excluded
`from the surface of the protein. Thereby the free
`energy required for denaturation is increased and
`the native structure of the protein is stabilized.5
`During the drying phase, the protein is stabilized
`by the water replacement mechanism and by the
`formation of a viscous glassy state. The water re-
`placement mechanism involves the formation of hy-
`drogen bonds between a protein and an excipient
`to satisfy the hydrogen-bonding requirement of po-
`lar groups on the protein surface.6 The formation
`of an amorphous viscous glass during freeze-drying
`and the corresponding extremely high viscosity
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`813
`
`Page 1
`
`

`
`814
`
`AL-HUSSEIN AND GIESELER
`
`also increase protein stability by retarding protein
`denaturation.7,8
`Disaccharides such as sucrose and trehalose are
`widely used as protein stabilizers, and these sug-
`ars have been extensively studied in the literature
`to investigate their stabilizing effect on proteins dur-
`ing freeze-drying. Several amino acids are frequently
`cited as being suitable excipients for freeze-drying of
`proteins. Glycine, for example, is widely used as crys-
`talline bulking agent in freeze-dried formulations,9
`whereas other amino acids such as lysine and argi-
`nine have been described as possible buffers in protein
`formulations.10 Arginine in combination with phos-
`phoric acid was reported to exert a stabilizing effect on
`lactate dehydrogenase (LDH) during freeze-drying.11
`In contrast to sugars, amino acids can also func-
`tion as buffers, and therefore, provide more choices/
`flexibility for the design of proteins formulations. His-
`tidine is one of the amino acids that can be used in
`protein formulations to function as both buffer and
`protein stabilizer. Histidine, which has three ioniza-
`tion sites on the molecule’s carboxyl, imidazole, and
`amino group with pK1 of 1.9, pK2 of 6.1, and pK3 of 9.1,
`has been used as a buffer especially in the pH range
`5–7.12
`Several studies have already referred to a stabi-
`lizing effect of histidine on proteins during freeze-
`drying. Although most of the stability studies deal
`with in-process instability of proteins, there were
`some (long-term) storage-stability studies. Osterberg
`et al.10 described the development of a stable freeze-
`dried formulation for recombinant factor VIII-SQ (r-
`VIII SQ) without the addition of albumin. The au-
`thors found that a combination of sucrose, nonionic
`surfactant (polysorbate 80), crystalline bulking agent
`(sodium chloride), and L-histidine preserve factor-
`VIII activity during freeze-drying and storage. It was
`also reported that L-histidine, besides functioning as a
`buffer, also had a stabilizing effect on r-VIII SQ during
`freeze-drying and storage. However, it is important to
`underline that the stabilizing effect of histidine was
`not studied in depth and not delineated and differen-
`tiated from the stabilizing effect of other stabilizers
`used in the same formulation. Cleland et al.13 used
`histidine as a buffer for the freeze-drying of a mon-
`oclonal antibody, rhuMAb HER2. The authors com-
`pared the stability profile of rhuMAb HER2 formu-
`lated at 25 mg/mL in either 5 mM succinate (pH 5)
`or 5 mM histidine (pH 6) in the presence of other
`excipients. They found that in the absence of sugar,
`a greater extent of aggregation was observed in the
`histidine formulation than in the succinate formu-
`lation. Chen et al.14 found that the increase of the
`histidine concentration from 4 to 6 mM reduced the
`soluble-aggregate levels of a human anti-IL8 mon-
`oclonal antibody (ABX-IL8) upon freeze-drying. The
`authors used multiple excipients systems consist-
`
`ing of glycine, glutamic acid, mannitol, and polysor-
`bate 20. Furthermore, the freeze-dried monoclonal
`antibody trastuzumab (Herceptin R(cid:2)) produced by
`F. Hoffmann-La Roche (Basel, Switzerland) is formu-
`lated with histidine.
`Overall, the reported studies about the stabilizing
`effect of histidine on proteins during freeze-drying are
`still limited, and all of these studies did not differen-
`tiate between the buffering effect of histidine and the
`other possible stabilizing effects of this amino acid on
`the proteins. Furthermore, literature does not provide
`data regarding the influence of histidine on proteins
`in the absence of other excipients that are regularly
`included in protein formulations. The presence of such
`excipients might complicate the identification of the
`effect of histidine on protein stability during freeze-
`drying.
`The scope of the present study was to investi-
`gate the influence of histidine on the stability of a
`model protein LDH for concentrations relevant for
`use as a buffer and as a stabilizer. Histidine buffer
`was compared with other common buffers (potassium
`phosphate and citrate). Furthermore, the ability to
`use histidine as a sole excipient in protein formu-
`lation was investigated. Moreover, the effect of pH
`and histidine concentration in the pre-freeze-dried
`bulk solution on both protein stability and the ele-
`gance of the final freeze-dried product was investi-
`gated. Because the amorphous state of the stabilizer
`is an essential property for the stabilization of pro-
`tein during freeze-drying, the physical state of his-
`tidine in freeze-dried samples with and without an-
`nealing was analyzed and correlated with the protein
`stability. LDH was selected for this study because of
`its well-documented labile nature and sensitivity to
`the stresses generated during freeze-drying.15 A com-
`parably low protein concentration of 15 :g/mL was
`employed to avoid protein self-protection, which is
`present at high concentrations.6,16
`
`MATERIALS
`l-Lactate dehydrogenase type II from rabbit mus-
`cle (11.4 mg protein/mL; 1150 units/mg) was used as
`aqueous suspension in ammonium sulfate and pur-
`chased from Sigma–Aldrich (Munich, Germany). L-
`Histidine, sodium pyruvate, and $-nicotinamide ade-
`nine dinucleotide (NADH) were also obtained from
`Sigma–Aldrich at analytical grade. Sucrose was ob-
`tained from Fluka Analytical (Buchs, Switzerland)
`and D-mannitol was purchased from Riedel-de Ha¨en
`(Seelze, Germany). Potassium dihydrogen phosphate
`(KH2PO4) and citric acid were obtained from Carl
`Roth (Karlsruhe, Germany) to prepare phosphate and
`citrate buffers, respectively. The pH of formulations
`containing histidine was adjusted to 4, 5, 6, and 7
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`DOI 10.1002/jps
`
`Page 2
`
`

`
`Table 1. Overview of the Studied Formulations and the Buffers in Each Part of this Study
`
`HISTIDINE STABILIZING EFFECTS ON LDH DURING FREEZE-DRYING
`
`815
`
`Studied Aspect
`
`Histidine as a buffer (comparison
`with other buffers)
`
`LDH
`(:g/mL)
`15
`
`Histidine as a stabilizer
`(comparison with other solutes)
`
`Histidine as buffer and stabilizer
`(histidine-concentration effect)
`
`Histidine as buffer and stabilizer
`(effect of solution pH)
`
`15
`
`15
`
`15
`
`15
`15
`15
`15
`
`15
`15
`15
`15
`15
`
`15
`15
`15
`15
`
`Buffer Concentration
`
`pH
`
`Nonbuffer Solute Concentration
`
`Citrate (10, 50, and 150 mM)
`
`Histidine (10, 50, and 150 mM)
`
`7.3 (Only buffer) Sucrose, S (10 mg/mL) Mannitol, M
`(10 mg/mL)
`7.3 (Only buffer) Sucrose, S (10 mg/mL) Mannitol, M
`(10 mg/mL)
`Phosphate (10, 50, and 150 mM) 7.3 (Only buffer) Sucrose, S (10 mg/mL) Mannitol, M
`(10 mg/mL)
`
`Potassium phosphate (10 mM)
`
`7.3
`
`Sucrose, S (100 mM)
`
`Histidine
`
`Histidine
`
`7.3
`7.3
`7.3
`7.3
`
`7.3
`7.3
`7.3
`7.3
`4
`
`5
`6
`7
`8
`
`Mannitol, M (100 mM)
`Histidine, H (100 mM)
`(Only buffer)
`Histidine (2 mg/mL)
`
`Histidine (5 mg/mL)
`Histidine (10 mg/mL)
`Histidine (20 mg/mL)
`Histidine (35 mg/mL)
`Histidine (20 mg/mL)
`
`Histidine (20 mg/mL)
`Histidine (20 mg/mL)
`Histidine (20 mg/mL)
`Histidine (20 mg/mL)
`
`with hydrochloric acid and to 8 with sodium hydrox-
`ide, whereas potassium hydroxide was used to adjust
`the pH in the formulations containing KH2PO4 as a
`buffer. Sodium hydroxide was used also to adjust the
`pH when citrate buffer was used. Potassium hydrox-
`ide, sodium hydroxide, and hydrochloric acid were
`purchased from Sigma–Aldrich. Vials (2 mL) were
`purchased from SCHOTT forma vitrum (M ¨ullheim,
`Germany). FluroTec R(cid:2) 13-mm stoppers were obtained
`from West Pharmaceutical Services (Eschweiler,
`Germany). To detect
`the possible pH shifts in
`the studied formulations during freezing, a univer-
`sal indicator solution was purchased from Sigma–
`Aldrich.
`The range of formulations that were freeze-dried
`and analyzed is illustrated in Table 1. The experi-
`ments were structured into four parts to evaluate the
`effect of histidine on LDH when applied as a buffer
`and as a stabilizer. First, histidine was used as a
`buffer in three different concentrations and compared
`with two other common buffer systems. The buffer
`formulations were also freeze-dried with addition of
`sucrose and mannitol. Second, histidine was used as
`a stabilizer and compared with sucrose and mannitol.
`The same buffer system was used for all combinations
`and was also freeze-dried without addition of stabiliz-
`ers for reference. In the third and fourth part, formu-
`lations containing only LHD and histidine in different
`concentrations and pH levels were investigated under
`consideration of the physical state.
`
`METHODS
`Preparation of Enzyme Solutions
`The LDH suspension was dialyzed with a spe-
`cial membrane (Spectra/Por R(cid:2) membrane, Spectrum
`Laboratories, Rancho Dominguez, California) with
`a molecular weight cut off (MWCO) of 12–14 kDa
`(molecular weight of LDH: 140 kDa). Dialysis was per-
`formed against potassium phosphate buffer (pH 7.3)
`at 5◦C overnight. The obtained enzyme solution was
`concentrated with an Amicon Ultra-15 centrifugal fil-
`ter device (MWCO 30 kDa; Millipore Corporation, Bil-
`lerica, Massachusetts) in a centrifuge (Minifuge RF,
`Heraeus Sepatech GmbH, Osterode, Germany). Pro-
`tein concentration was determined spectrophotomet-
`rically at 280 nm. Aliquots of dialyzed LDH and ex-
`cipients solution were mixed in glass vials to obtain
`0.5-mL samples with a final concentration of 15 :g/
`mL enzyme.
`
`Freeze-Drying Process Conditions
`Individual formulation containing LDH (0.5 mL) was
`filled into 2-mL vials, and the vials were subsequently
`semistoppered. Freeze-drying experiments were per-
`formed using a FTS LyostarTM II (SP Scientific, Gar-
`diner, New York). Samples were loaded onto the
`shelves at room temperature, frozen using a 1◦C/min
`shelf cooling rate down to–40◦C, and maintained at
`this temperature for 1 h. Annealing (when applied)
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`Page 3
`
`

`
`816
`
`AL-HUSSEIN AND GIESELER
`
`Table 2. Obtained Glass Transition Temperature (T (cid:3)
`Studied Formulations
`
`g) for the
`
`Formulationsa
`Only histidine formulationsc
`
`Sucrose formulationsd
`
`Mannitol formulationsd
`
`pH 4
`pH 5
`pH 6
`pH 7
`pH 8
`In phosphate buffer
`In citrate buffer
`In histidine buffer
`In phosphate buffer
`In citrate buffer
`In histidine buffer
`
`g (◦C)b
`T(cid:3)
`–49.66
`–49.12
`–40.80
`–35.50
`–36.85
`–34.07
`–33.25
`–30.80
`–32.35
`–33.27
`–38.58
`
`aLDH concentration in all studied formulations is 15 :g/mL.
`bMeasurements were performed in triplicate (n = 3). All standard
`deviations were <1.4◦C.
`cHistidine concentration is 20 mg/mL.
`dpH 7.3.
`
`was conducted by ramping the shelf temperature from
`−40◦C to −20◦C at 1◦C/min and keeping this shelf
`temperature for 4 h for thermal treatment. Then, the
`product was frozen back to −40◦C and kept at this
`temperature for 1 h. For all formulations, primary
`drying was performed by controlling the shelf temper-
`ature at −30◦C and the chamber pressure at 80 mTorr
`for 25 h. The selected shelf temperatures for anneal-
`ing and primary drying were selected on the basis of
`the glass transition temperatures (T(cid:3)
`g) of the studied
`formulations, which were determined by differential
`scanning calorimetry (DSC). It is well known that an-
`nealing should be carried out at temperature above
`T(cid:3)
`g of the formulation.3 The selected annealing tem-
`perature in this study (–20◦C) was higher than the
`measured glass transition temperatures of all studied
`formulation. The glass transition temperatures (T(cid:3)
`g)
`of the studied formulations are reported in Table 2.
`The conditions used are very conservative for conven-
`tional freeze-drying, and the rationale behind the use
`of such experimental parameters is (1) to avoid any
`impact of the primary drying phase on the physico-
`chemical state of the mixture and (2) to verify that
`differences in protein stability between different for-
`mulations did not arise from differences in drying
`conditions.
`Lastly, secondary drying was carried out at the
`same chamber pressure applied during primary
`drying but increasing the shelf temperature to +40◦C
`at a ramp rate of 0.1◦C/min and maintaining this
`temperature for 4 h. The shelf temperature and dura-
`tion of secondary drying were selected to obtain final
`products with relatively low and comparable residual
`moisture content to ensure that differences in protein
`stability between different formulations are not
`attributed to different residual moisture contents.
`Throughout this study, product temperatures during
`the cycle were measured using calibrated 36-gauge
`
`from
`thermocouples
`copper/constantan
`T-Type
`Omega (Omega Engineering, Stamford, Connecti-
`cut). Each thermocouple was introduced through a
`stopper and positioned bottom-center in the vial to
`achieve both representative temperature monitoring
`as well as accurate endpoint detection of the time
`point when no ice was left in the product.
`
`Assay to Assess the Enzymatic Activity
`Enzymatic-activity recovery was used in this study
`as an indicator for the functional stability of LDH.
`LDH catalyzes the interconversion of pyruvate to lac-
`tate with concomitant interconversion of NADH to
`NAD+. The decomposition of NADH was measured
`by the decrease in absorption at 340 nm. A 10-mm
`quartz cuvette was placed into a Lambda 25-UV/Vis
`spectrometer (PerkinElmer, Rodgau, Germany) con-
`nected to a computer system (WinLab V 5.0 software,
`PerkinElmer). The rate of absorbance decrease is di-
`rectly proportional to LDH activity in the sample. Ac-
`tivity was measured before freeze-drying and after re-
`constitution of the freeze-dried product. The enzyme
`activity was defined as 100% in the solution prior to
`freezing, and the remaining activity of LDH in the
`reconstituted freeze-dried samples was expressed as
`percentage of the original activity before freezing. All
`assays were performed in triplicate. Both the mean
`value and the standard deviation were calculated.
`The variation coefficient of this assay ranged between
`4.3% and 4.7%.
`
`Turbidity Measurements
`The turbidity of solutions was measured to quantify
`protein denaturation that led to the formation of in-
`soluble protein aggregates. Turbidity values were ob-
`tained by UV-spectroscopy measurements at 350 nm.
`
`Fourier Transform Infrared Spectroscopy
`Fourier transform infrared spectroscopy (FTIR) was
`employed to evaluate potential changes in pro-
`tein secondary structure after freeze-drying. The
`evaluation was executed through comparison of
`second-derivative spectrum of untreated LDH be-
`fore freeze-drying with second-derivative spectra of
`LDH obtained after the reconstitution of
`freeze-
`dried samples. FTIR spectra of samples containing
`LDH were obtained using a Nicolet Magna IR 550
`FTIR spectrometer (Thermo Fisher Scientific Inc.,
`Waltham, Massachusetts). The apparatus was con-
`stantly purged with dry air. Samples were measured
`in a temperature-controlled CaF2 window with a fixed
`sample-layer-thickness of 5.6 :m. The water spec-
`trum was subtracted from the sample spectrum us-
`ing the Nicolet Omnic software (Thermo Scientific,
`Waltham, MA, USA). A subtraction of the background
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`DOI 10.1002/jps
`
`Page 4
`
`

`
`HISTIDINE STABILIZING EFFECTS ON LDH DURING FREEZE-DRYING
`
`817
`
`signal of the excipients was performed manually. The
`amide I band range between 1580 and 1720 cm−1 was
`isolated from the spectrum and a baseline correction
`was executed. Individual peak positions were identi-
`fied for the second-derivative spectrum of this range.
`
`Differential Scanning Calorimetry
`Thermal analysis was conducted using a DSC822e
`(Mettler Toledo, Greifensee, Switzerland) to evalu-
`ate the thermal properties of the solutions and the
`freeze-dried cakes. The samples were prepared in
`40-:L Al pans in a glove box (relative humidity <1%)
`and hermetically sealed at room temperature. Pans
`were weighed before (empty) and after filling on a
`calibrated microbalance (AT 261 Delta Range, Met-
`tler Toledo) to determine the exact weight of each in-
`dividual sample. After placing the pans in the DSC
`measurement cell, nitrogen was used for purging and
`drying (30 and 150 mL/min, respectively). The freeze-
`dried samples were heated from room temperature to
`300◦C at a heating rate of 5◦C/min, whereas the liquid
`samples were cooled from room temperature to −80◦C
`at cooling rate 5◦C/min and held at this temperature
`for 5 min before being heated to 5◦C at a heating rate
`of 2◦C/min.
`All thermal events including the determination of
`the glass transition temperature (T(cid:3)
`g) were evaluated
`using the Mettler STARe Software V 9.01 (Mettler
`Toledo).
`
`X-Ray Powder Diffraction
`To characterize the crystallinity of the freeze-dried
`products, samples were examined by X-ray powder
`diffraction (XRPD) using a Philips X’pert MPD with
`Cu K" radiation at 40 kV/40 mA (Philips Analytical
`Technology, Munich, Germany). Powder samples were
`filled into an Al sample holder and compressed using
`a simple cover glass. All scans were measured in the
`range 2θ = 0.5◦–40◦, with a step size of 0.02◦ (time
`per step = 1 s).
`
`Residual Moisture Content (Karl-Fischer Titration)
`The residual moisture content in the freeze-dried
`samples was determined by Karl-Fischer titration
`using a Metrohm Coulometric KF titrator (831 KF
`Coulometer, Metrohm, Filderstadt, Germany) with an
`oven system. All measurements were performed in
`triplicate at an oven temperature of 120◦C.
`
`RESULTS AND DISCUSSION
`Residual Moisture Contents
`The obtained residual moisture contents in the freeze-
`dried cakes were comparable (about 1%), and no
`substantial differences were observed in the water
`
`content between the freeze-dried samples. Therefore,
`the different protein stabilities in the studied formu-
`lations (see below) did not result from different resid-
`ual moisture contents.
`
`Comparison of Histidine as a Buffer System with Citrate
`and Phosphate Buffers
`Lactate dehydrogenase was freeze-dried without an-
`nealing in three different buffers (histidine, citrate,
`and potassium phosphate) at three different buffer
`concentrations (10, 50, and 150 mM). The pH for all
`buffers was adjusted to 7.3 (see Table 1). For this part
`of the study, LDH was freeze-dried either in formula-
`tions containing solely the studied buffers or in excip-
`ient systems containing sucrose or mannitol (10 mg/
`mL) in addition to the buffer. The rationale for this
`combination of experiments is to obtain comprehen-
`sive information about the effect of histidine buffer
`on protein stability in different environments, that is,
`when no other solutes are present in the formulation,
`compared with inclusion of common excipients such
`as sucrose and mannitol. Figure 1 illustrates the ac-
`tivity recovery of LDH after the reconstitution of the
`studied formulations. Irrespective of the formulation
`composition, remaining activities of LDH in histidine
`buffer were higher than those observed with citrate
`or phosphate buffer at all buffer concentrations. The
`addition of sucrose or mannitol to the formulations
`resulted in a significant increase in the protein ac-
`tivity, and the higher residual activities of LDH were
`obtained in sucrose formulations. However, remain-
`ing activity of LDH in histidine buffer compared with
`that in the other buffers was also higher in the pres-
`ence of sucrose or mannitol, indicating a higher de-
`gree of protein stability in the presence of histidine. It
`is noteworthy that remaining activity of LDH is lower
`in phosphate buffer than in citrate and histidine, and
`this difference is very distinct in formulations con-
`taining only protein and buffer, where protein activ-
`ity was dramatically improved in citrate and histidine
`samples. It is well known that pH shifts can occur dur-
`ing freezing because of the selective crystallization of
`the buffer. This change of pH might affect protein
`stability and result in protein denaturation. The pos-
`sible pH changes in the studied formulations were in-
`vestigated using a universal pH indicator, which was
`added to the formulations prior to freezing. The pH
`shifts were observed in all formulations formulated
`with phosphate buffer at concentrations of 50 and
`150 mM, whereas no pH shifts were observed with
`citrate or histidine formulations. Note that the selec-
`tive crystallization of phosphate buffer during freez-
`ing, particularly at elevated concentrations, was also
`reported by Pikal.17
`As a side note, mannitol is rarely used as protein
`stabilizer during freeze-drying because of its strong
`
`DOI 10.1002/jps
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`Page 5
`
`

`
`818
`
`AL-HUSSEIN AND GIESELER
`
`Figure 1. Activity recovery of freeze-dried LDH (15 :g/mL) in different buffers (pH 7.3) at
`10, 50, and 150 mM. Dashed columns, only buffer formulations; empty columns, sucrose formu-
`lations; and filled columns, mannitol formulations. C, citrate; H, histidine; and P, potassium
`phosphate. Solid lines in each bar represent standard deviations.
`
`tendency to crystallize, and therefore, mannitol is nor-
`mally used as crystalline bulking agent.2,4 However,
`in the presented results, mannitol also showed a sta-
`bilizing effect on LDH, which might be ascribed to
`the fact that freeze-drying was performed without
`annealing and therefore, a fraction of mannitol re-
`mained amorphous and exerted the stabilizing effect.
`To gain further insight into protein denaturation
`mechanisms in the studied formulations, the turbid-
`ity of protein solutions was measured after reconsti-
`tution of the freeze-dried samples. Turbidity values
`of formulations to which only buffer was added are
`listed in Table 3. Note that very low turbidity results
`(between 0.002 and 0.004) were found in the solutions
`prior to freeze-drying. Turbidity values of freeze-dried
`histidine buffer formulations were lower compared
`with freeze-dried formulations of the other buffers at
`all used concentrations. In contrast, the turbidity val-
`ues of the formulations containing phosphate buffer
`were significantly elevated compared with those ob-
`tained with citrate and histidine buffers. The results,
`therefore, indicate that protein denaturation mani-
`fested by the formation of insoluble aggregates oc-
`curred to a lower degree in formulations containing
`histidine. Protein denaturation might be manifested
`also by the formation of soluble aggregates, but these
`aggregates play no role in the turbidity of a protein so-
`lution and therefore, they are neglected by measuring
`the turbidity of the protein solution. Note that turbid-
`
`Table 3. Turbidity Values for LDH at 15 :g/mL of Reconstituted
`Samples Freeze-Dried in Different Buffers
`
`Formulationsa
`C10 mM
`H10 mM
`P10 mM
`C50 mM
`H50 mM
`P50 mM
`C150 mM
`H150 mM
`P150 mM
`
`Turbidity Aλ350
`(Optical Density at 350 nm)b
`
`0.015
`0.009
`0.032
`0.022
`0.010
`0.038
`0.010
`0.007
`0.043
`
`aC, citrate; H, histidine; P, phosphate.
`bMeasurements were performed in triplicate (n = 3). All standard
`deviations were <0.005.
`
`ity values are consistent with the results of the activ-
`ity assay where greater loss of LDH activity occurred
`in formulations that did not contain histidine. Note
`that higher concentrations of potassium phosphate
`buffer were associated with both decreased activity
`recovery and increased turbidity in the reconstituted
`samples.
`Further insight into potential changes in secondary
`protein structure of LDH in the studied formulations
`was obtained from FTIR measurements. The amide I
`region (1600–1700 cm−1) is most commonly employed
`to analyze the secondary structure of proteins.18 How-
`ever, the original spectrum of amide I is usually
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 3, MARCH 2013
`
`DOI 10.1002/jps
`
`Page 6
`
`

`
`HISTIDINE STABILIZING EFFECTS ON LDH DURING FREEZE-DRYING
`
`819
`
`Figure 2. Amide I band peak (dotted line) and second-derivative spectrum (solid line) of LDH.
`Assignment of peak positions to individual secondary structures is given in the figure.
`
`featureless, and the individual underlying com-
`ponents of secondary structure cannot be visual-
`ized. Therefore, the second-derivative spectrum of
`the amide I band is used to delineate overlap-
`ping secondary-structure components in the original
`spectrum.19 Figure 2 depicts the second-derivative
`spectrum of buffered LDH solution prior to freeze-
`drying, with the assignments of peaks to secondary-
`structure components.20 According to literature,21,22
`peak positions were assigned to

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