throbber
European Journal of Pharmaceutical Sciences 8 (1999) 301–308
`
`Physical state of L-histidine after freeze-drying and long-term storage

`a
`Thomas Osterberg , Tommy Wadsten


`Pharmaceutical and Analytical R&D, Astra Pain Control AB, S-15185 Sodertalje, Sweden
`bDev & Res Wadsten AB, S-11327 Stockholm, Sweden
`
`b
`
`a
`
`Received 27 November 1998; received in revised form 2 March 1999; accepted 18 March 1999
`
`Abstract
`
`Liquid samples of L-histidine of varying pH values and mixed with salt, metal ions, polysorbate 80 and sucrose have been analysed by
`differential scanning calorimetry to evaluate the influence of these additives on the glass transition temperature and crystallisation of
`L-histidine during freezing and thawing. L-Histidine solutions of varying pH were freeze-dried with and without a thermal cycle and the
`physical state of the freeze-dried cakes, following long-term storage, were studied by powder X-ray diffraction. Amorphous L-histidine
`crystallised when it was exposed to moisture, and the identity of the crystalline materials is reported. The crystallisation of L-histidine
`during freezing and thawing is dependent on the pH of the solution and is shown to be at a minimum at pH 6, which coincides with the
`pK of the imidazoline function. Sucrose inhibited the crystallisation of L-histidine during thawing, while sodium chloride or polysorbate
`a
`21
`21
`80 did not. The addition of metal ions (Ca
`and Mg
`) up to 10% (w/w) did not depress the glass transition temperature significantly,
`21
`while the addition of Zn
`increased it. The physical state of L-histidine after freeze-drying is shown to be dependent on both the pH of
`the solution and the freezing cycle. The risk of crystallisation of amorphous L-histidine is low if the freeze-dried material is protected from
`moisture.
`1999 Published by Elsevier Science B.V. All rights reserved.
`
`Keywords: Amorphous; Crystallisation; Freeze-drying; Glass transition; L-Histidine; Metal ions
`
`1. Introduction
`
`Protein drugs are generally chemically and physically
`unstable in solution and freeze-drying is frequently used to
`obtain an acceptable shelf life (MacKenzie, 1966, 1977;
`Pikal, 1990). Sugars and/or amino acids are often included
`in the formulation to prevent inactivation during freeze-
`drying and to stabilise the protein during long-term
`storage. Sugars and amino acids protect the protein by
`preferential exclusion during freezing and by glass forma-
`tion and /or by functioning as a water substitute in the
`dried state (Carpenter and Crowe, 1989; Franks et al.,
`1991; Arakawa et al., 1992). In contrast to sugars, amino
`acids, in addition to their stabilising properties, may also
`function as buffers. L-Histidine has recently been shown to
`function as both buffer and stabiliser
`in freeze-dried

`formulations of recombinant factor VIII (Osterberg et al.,
`1997) and recombinant factor IX (Bush et al., 1998). It is a
`basic amino acid often found at the active site in enzymes
`and in the coordination of metal ions in metalloproteins.
`The specific properties of L-histidine reside in the im-
`idazoline function, which possesses both basicity and p-
`

`E-mail address: thomas.osterberg@eu.pnu.com (T. Osterberg)
`
`electron acceptor capability (Sundberg and Martin, 1974).
`The imidazoline function confers good buffer capacity in
`the pH range 5–7 (Fig. 1), which is often a suitable pH
`range for many protein drugs. L-Histidine forms strong
`21
`21
`complexes with certain metal ions such as Cu , Zn
`and
`21
`1
`1
`Fe
`, but simple salts with the alkali metals (Na , K ,
`21
`21
`Ca
`and Ba
`) (Greenstein and Winitz, 1961). The latter
`fact is especially important in the formulation of protein
`drugs that require free calcium ions in the buffer for
`stability reasons (e.g. factor VIII). Trace levels of metal
`ions such as copper and iron are often present in buffer
`salts. These ions can often facilitate the oxidation of
`proteins (Lamfrom and Nielsen, 1970; Shihong et al.,
`1993). Since L-histidine forms strong complexes with these
`ions, it may also function as an antioxidant. The solubility
`of L-histidine in water is 41.9 mg/ml at 258C and is
`sufficient for proper buffering and to allow it to function as
`a non-crystallising (amorphous) stabiliser for many protein
`drugs. In this study the freezing/thawing behaviour of
`L-histidine of varying pH in the presence of sodium
`chloride, metal ions and sucrose was studied by means of
`differential scanning calorimetry (DSC). L-Histidine solu-
`tions of varying pH were freeze-dried with and without a
`thermal cycle. The freeze-dried cakes were examined with
`
`0928-0987/99/$ – see front matter
`PII: S0928-0987( 99 )00028-7
`
`1999 Published by Elsevier Science B.V. All rights reserved.
`
`CFAD Exhibit 1030
`
`1
`
`(cid:211)
`(cid:211)
`

`

`302
`

`T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`The solutions were sterile-filtered and 1.0 ml was
`dispensed into glass vials. About 10 vials of each formula-
`2
`tion were prepared. A pilot freeze-dryer with a 1.2-m
`shelf area (Edward Kniese, Marburg, Germany) was used.
`The samples were frozen on the shelves (from 0 to 2608C
`in 1 h). The thermal cycle was 260 to 2358C in 1 h,
`maintaining 2358C for 4 h and cooling to 2608 in 1 h.
`Primary drying was carried out at a shelf temperature of
`about 2208C and the pressure was adjusted to give a
`product temperature of about 2408C. On completion of the
`primary drying, the shelf temperature was raised to 408C
`over 10 h. The completion of the primary and secondary
`drying was determined by a pressure increase test. The
`vials were stoppered under a vacuum in the freeze-drier.
`The freeze-drying process is illustrated schematically in
`Fig. 2.
`
`2.2.2. Powder X-ray diffraction
`Powder X-ray diffraction data were obtained with a

`powder diffractometer (XPert, Philips, The Netherlands).
`Data were collected at room temperature from 3 to 358, 2u,
`the step size was 0.028 and the count time was 0.5 s.
`Powder samples were prepared as thin layers on glass or
`aluminium specimen holders. The samples were first
`analysed at ambient
`temperature and then exposed to
`moisture from 20 to 80% RH at room temperature in the
`diffractometer by using a humidity control unit
`(also
`Philips-made).
`
`2.2.3. Water sorption/desorption
`The water sorption/desorption profile was determined
`with an isothermal dynamic water uptake instrument (MB-
`300W, VTI Corporation, USA). The sample was dried
`under nitrogen at 608C. The temperature was thereafter set
`to 358C and the relative humidity was increased from 0 to
`80% for sorption and decreased from 80 to 5% for
`desorption in steps of 5%. The initial sample mass was
`about 7 mg and the equilibrium criterion was less than
`0.005 mg mass gain per 3 min. Two vials of each
`formulation (pH 5, 6 and 8) were analysed.
`
`Fig. 2. Plot of process variables during freeze-drying.
`
`Fig. 1. Simulated titration curve of L-histidine showing the major
`electrical forms at varying pH.
`
`powder X-ray diffraction after long term-storage and after
`moisture exposure. The exposure to moisture induced
`crystallisation and the identity of the crystalline materials
`is reported.
`
`2. Experimental
`
`2.1. Materials
`
`L-Histidine used for the freeze-drying experiments con-
`formed to the requirements laid down in DAB and USP.
`L-Histidine (SigmaUltra), L-histidine monohydrochloride
`monohydrate, sucrose (SigmaUltra), and sodium chloride
`(SigmaUltra), used for DSC studies and as references for
`the powder X-ray experiments, were
`from Sigma
`(Sweden). Magnesium chloride (ACS reagent grade, ICN
`Biomedicals), calcium chloride dihydrate (ACS reagent
`grade, Acros) and Tween 80 (Polysorbate 80) were
`obtained from Chemicon (Sweden). Zinc chloride (Ph.
`Eur) came from KEBO Lab (Sweden). The sterile filter
`used was a 0.22-mm, Millex GV (Millipore, Sweden). The
`containers used were of type 1 (Ph. Eur). Bottles were
`closed with a bromobutyl rubber stopper and sealed with
`an aluminium seal. Water for injections, hydrochloric acid
`and sodium hydroxide were of pharmacopoeial grade.
`
`2.2. Methods
`
`2.2.1. Preparation of freeze-dried samples
`L-Histidine was dissolved in water for injections (15
`mg/ml) and the pH was adjusted to pH 4, 5, 6, and 7 with
`hydrochloric acid and to pH 8 with sodium hydroxide.
`
`2
`
`

`

`¨T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`303
`
`2.2.4. Thermal analysis
`
`2.2.4.1. Preparation of samples for thermal analysis
`for
`L-Histidine, 30 mg/ml, was dissolved in water
`injections and adjusted with hydrochloric acid or sodium
`hydroxide to the different pH values. Mixtures of L-
`histidine and sucrose were prepared by mixing L-histidine
`(30 mg/ml) and sucrose (30 mg/ml) in different volume
`ratios. The solutions were dispensed (about 30 ml) in a
`50-ml aluminium pan without a lid. An empty pan was
`used as reference.
`
`2.2.4.2. DSC
`A differential scanning calorimeter, DSC6200, with an
`EXTAR6000 workstation (Seiko Instruments, Japan), was
`used. Cyclohexane (SigmaUltra) and indium (Laboratory
`of the Government Chemist, UK) were used to calibrate
`the instrument. The oven was cooled with liquid nitrogen.
`The sample was cooled to 21108C at a rate of 108C/ min.
`After an equilibration period of 2 min the sample was
`heated to 2208C at 108C/min. The sample was cooled
`again to 21108C, equilibrated for 2 min and subsequently
`reheated to 08C at 108C/min. The sample was inspected
`under a magnifying glass or stereomicroscope directly after
`analysis when some ice was still melting to observe
`possible crystallisation. Runs were also conducted with a
`glass lid on the oven so that crystallisation could be
`observed visually. Reported glass transition temperatures
`are midpoint values (mean value from two runs) from the
`second scan, and were determined with help of
`the
`software and the derivative of the DSC signal.
`
`3. Results and discussion
`
`The selection of buffer for a protein formulation is very
`important and several factors have to be considered. The
`buffer must have low local and systemic toxicity and be
`compatible with the active protein and other essential
`ingredients (e.g. metal ions). It must also be chemically
`stable and the pK should preferably be close to the
`a
`formulation pH in order to give good buffer capacity. The
`latter requirement also implies that the extent of ionisation
`of the buffer varies considerably around the formulation
`pH. This also applies to drug compounds with a pK close
`a
`to the pH of the formulation. For example, a compound
`with a pK of 7.0 is 50% in the ionised state at pH 7.0, and
`a
`a decrease by one pH unit increases the ionisation to 90%.
`Thus, for compounds with a pK near the formulation pH,
`a
`we are dealing with different
`ratios of charged and
`uncharged forms depending on relatively small changes in
`the formulation pH. This is in contrast to non-ionisable
`compounds (such as sucrose and mannitol) or compounds
`with a pK a long way from the formulation pH. Histidine
`a
`has three ionisable functions: the carboxyl group (pK 5
`1
`1.9),
`the imidazole nitrogen (pK 56.1) and the amino
`2
`
`nitrogen (pK 59.1) at 258C and 0.15 ionic strength
`3
`(Sundberg and Martin, 1974). A theoretical titration curve
`with the major electrical forms of L-histidine is shown in
`Fig. 1. Since the pK is close to the pH range often used in
`2
`the formulation of protein drugs, the net electrical charge
`of the imidazoline moiety as a function of pH is of
`fundamental importance for its physicochemical properties
`and its behaviour during freezing and in the dried state.
`
`3.1. DSC
`
`The thermal behaviour of L-histidine during freezing and
`thawing has recently been studied by (Chang and Randall,
`1992), who reported that L-histidine (10 mg/ml) remains
`amorphous during freezing, with a T value of 2328C.
`9
`g
`The T of L-histidine (30 mg/ml) measured in the present
`9
`g
`study was 231.58C, although L-histidine crystallised dur-
`ing thawing at about 2108C. Since this was in contrast to
`the findings reported by Chang and Randall (1992), a
`sample of 10 mg/ml and rapid freezing was also investi-
`gated. The T was 2328C, although crystallisation took
`9
`g
`place at about the same temperature as for the sample
`consisting of 30 mg/ml. However, since the crystallisation
`takes place during the softening/melting of the ice, the
`exotherm is obscured. An exotherm was observed at about
`2108C in the sample of 30 mg/ml but not in the sample of
`10 mg/ml. The crystallisation could also be observed
`visually if a glass lid was used on the calorimeter oven.
`The crystallisation takes place mainly on the surface of the
`sample solution and the crystals have a flat, round appear-
`ance. This type of crystals was also observed on the
`surface of some of the freeze-dried samples. The thermo-
`gram from the pH 4 sample showed crystallisation between
`230 and 2208C. Crystallisation was also observed visual-
`ly in the sample cup in this temperature range and also in
`the sample cup directly after thawing. No crystallisation of
`L-histidine during thawing was observed in samples in the
`pH range 5.5–6.0. The pH 6.5 samples showed one small
`crystal. Thus, the findings from this study indicate that the
`tendency for crystallisation is at a minimum between 5.5
`and 6.5. Crystallisation of L-histidine was also observed in
`the samples containing sodium chloride. Thus, the reduced
`crystallisation of L-histidine between pH 5.5 and 6.5 is
`most
`likely due to the ionisation of
`the imidazoline
`function, since the addition of sodium chloride (up to 0.6
`M and unadjusted pH) did not inhibit the crystallisation.
`The thermal analysis of L-histidine solutions of varying pH
`showed a sigmoidal relationship between pH and T (Fig.
`9
`g
`3). The T at pH 5 was about 108C lower than at pH 7.
`9
`g
`The effect of pH on T can be explained by the plasticis-
`9
`g
`ing effect of ions (from pH adjustment with HCl or NaOH)
`in the freeze-concentrated L-histidine glass and/or a lower
`T of L-histidine with an ionised imidazole moiety. It is
`9
`g
`well known that salts decrease T
`and the collapse
`9
`g
`temperature of freeze-concentrated amorphous materials.
`The T and the collapse temperature of amorphous materi-
`9
`g
`
`3
`
`

`

`304
`

`T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`Fig. 3. Dependence of T during the second heating of frozen aqueous
`9
`g
`L-histidine solutions on the pH and on the addition of sodium chloride.
`
`als are generally closely related (MacKenzie, 1977). The
`of
`the sucrose/ sodium chloride system has been
`9T
`g
`described by (MacKenzie, 1985). The addition of sodium
`chloride to sucrose in 1:9 and 2:8 (w/w) ratios depressed
`the T by about 10 and 218C, respectively. If one assumes
`9
`g
`that sodium and chloride ions depress the collapse tem-
`perature equally, the chloride ions alone would depress the
`9T by about 5 and 108C, respectively,
`in the sucrose /
`g
`sodium chloride system above. L-Histidine (30 mg/ ml)
`adjusted to pH 6 with hydrochloric acid contains about 3.4
`mg of chloride ions. The L-histidine/chloride ion ratio is
`thus approximately 10:1 (w/w). If one further assumes that
`sodium chloride depresses the T of sucrose and L-his-
`9
`g
`tidine equally, the chloride ions would depress the T of
`9
`g
`L-histidine (pH 6) by about 108C. The measured T was
`9
`g
`about 88C lower at pH 6 compared to the unadjusted state,
`which is quite close to the value estimated from the
`sucrose/sodium chloride system. However, it is not pos-
`sible to establish the T of L-histidine at pH 6 per se due to
`9
`g
`the presence of the pH adjuster, although the reasoning
`above indicates that
`the ionisation of the imidazoline
`function does not change the T dramatically. Since L-
`9
`g
`histidine was shown to crystallise during thawing in the
`important pH range 6.5–8 it was important to investigate
`whether the addition of a non-crystallising excipient could
`function as an inhibitor. Sucrose was selected since it is
`often used as a non-crystallising excipient in freeze-dried
`formulations of protein drugs. The T of L-histidine/ suc-
`9
`g
`rose mixtures of varying pH is shown in Fig. 4. The T of
`9
`g
`mixtures of two non-crystallising compounds can generally
`be approximated using the Fox equation (Fox, 1950),
`
`Fig. 4. Dependence of T during the second heating of frozen aqueous
`9
`g
`L-histidine–sucrose solutions on the weight to weight ratio of L-histidine–
`sucrose and pH.
`
`are the
`is the new T and W , T , W and T
`where T
`g
`1
`g 1
`2
`g 12
`g 2
`weight fractions and the T of the individual compounds.
`g
`The T values measured in this study were 231.58C for
`9
`g
`L-histidine (30 mg /ml) and 232.38C for sucrose (30
`mg/ml). Thus, the estimated T of any L-histidine–sucrose
`9
`g
`mixture will not deviate much from 2328C.
`It was,
`therefore, interesting to note that the T of all the mixtures
`9
`g
`(unadjusted pH) was about 3–48C higher compared to the
`T values estimated from the Fox equation. The T at pH
`9
`9
`g
`g
`7 was essentially unchanged for the samples with 33 and
`50% (w /w) L-histidine. The samples with 33 and 50%
`(w/w) L-histidine of pH 6 showed a T above 2358C
`9
`g
`which is a reasonable T from a practical point of view.
`9
`g
`The T of pure L-histidine at pH 6 was 2408C. A T at or
`9
`9
`g
`g
`below 2408C is generally considered to be too low since
`the sublimation of ice is very slow at this temperature. As
`a rule of thumb, the time for the primary drying is halved
`if the product temperature is increased by 6–78C. Thus, the
`increased T
`of the L-histidine/sucrose mixtures is of
`9
`g
`economic and practical
`importance. Another important
`observation was that the addition of sucrose abolished the
`crystallisation of L-histidine. The reduced tendency for
`crystallisation of L-histidine is very important
`in the
`formulation design, since even if the product temperature
`never reaches the devitrification temperature during normal
`operating conditions, accidental over-heating might occur
`in the freeze-dryer due to technical failures.
`
`3.2. Addition of metal ions and polysorbate 80
`
`1/T 5 W /T 1 W /T
`g 12
`1
`g 1
`2
`
`g 2
`
`Some proteins like factor VIII require divalent metal
`
`4
`
`

`

`¨T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`305
`
`21
`
`) in the formulation buffer for stability
`ions (e.g. Ca
`reasons. Proteins that are therapeutically active at very low
`concentrations usually require the addition of non-ionic
`surfactants (e.g. polysorbate 80) in order to retard the
`surface adsorption of the protein during manufacture and
`in the final packaging. It was,
`therefore, of interest
`to
`21
`21
`investigate whether the divalent metal ions, Ca
`, Mg
`21
`and Zn
`(added as chloride salts) or polysorbate 80, could
`retard the crystallisation and how these ingredients in-
`fluenced the T of L-histidine. Sucrose was included in
`9
`g
`order to study how an uncharged compound interacts with
`these metal
`ions. The crystallisation of L-histidine (30
`mg/ml unadjusted pH) was largely unaffected by the
`presence of calcium chloride (up to 10 mg/ml), but was
`completely retarded at 20 mg/ml. The addition of poly-
`sorbate 80, 0.2 mg/ ml, did not inhibit the crystallisation.
`21
`21
`21
`The effect of Ca
`, Mg
`and Zn
`on the T of L-
`9
`g
`histidine and sucrose is shown in Fig. 5. The addition of
`metal ions to sucrose showed an expected concentration-
`21
`dependent depression of the T . Mg
`depressed the T of
`9
`9
`g
`g
`21
`21
`sucrose most, followed by Ca
`and Zn . However, in
`21
`21
`contrast to sucrose, the addition of Ca
`and Mg
`to
`L-histidine showed a surprisingly small depression of T of
`9
`g
`up to 10% (w/w) ratio. Another unexpected finding was
`21
`that the addition of Zn
`increased the T considerably.
`9
`g
`21
`21
`and Mg
`The small depression of T by Ca
`up to 10%
`9
`g
`(w/w) ratio indicates that L-histidine interacts with these
`metal
`ions in the freeze-concentrated phase. It
`is well
`21
`known that L-histidine forms a strong complex with Zn ,
`and this probably explains the increase in the T . The
`9
`g
`
`21
`
`21
`
`of L-
`of the T
`and Mg
`small depression by Ca
`9
`g
`histidine is of considerable importance in the formulation
`of proteins that require metal
`ions for stability. If L-
`histidine is used as buffer/stabiliser in formulations con-
`21
`21
`taining Ca
`and Mg , the T is essentially unchanged,
`9
`g
`the T
`whereas if sucrose is used,
`can be depressed
`9
`g
`considerably.
`
`3.3. Freeze-drying
`
`The freezing step is very important since the internal
`structure of the product
`is determined by this step. A
`thermal cycle (Fig. 2) is sometimes necessary to maximise
`the crystallisation of water and/or to promote the crys-
`tallisation of bulking agents such as sodium chloride or
`mannitol. Since a thermal cycle might promote crystallisa-
`tion of ingredients (e.g. L-histidine) that are to remain
`amorphous, the L-histidine samples were freeze-dried with
`and without a thermal cycle. Freeze-drying of L-histidine
`from solutions having a pH in the range 4–8 showed that
`L-histidine has a rather low tendency to crystallise during
`freeze-drying (Table 1). The samples freeze-dried at pH 8
`crystallised readily, but only when they were freeze-dried
`with a thermal cycle. The samples of pH 7 and 8, freeze-
`dried without a thermal cycle, and the sample with pH 7,
`freeze-dried with a thermal cycle, showed very thin crystal
`clusters (1–2 mm in diameter) on the surface of the cakes.
`All samples showed good cake structures, with the excep-
`tion of the pH 4 sample, which collapsed to a transparent
`skin during freeze-drying. Examination of the samples
`with a polarising microscope showed that the pH 8 sample
`freeze-dried with a thermal cycle had a crystalline struc-
`ture, while the other samples had an amorphous appear-
`ance. The crystallisation of L-histidine at pH 8 during
`freeze-drying might be explained by the fact
`that
`the
`imidazoline moiety is almost completely un-ionised at this
`pH and that crystallisation is generally facilitated if only
`one molecular species is present. At pH 6 there is a 1:1
`molar ratio of the charged and the uncharged imidazole
`moiety and it is suggested that this ratio is optimum for the
`inhibition of crystallisation during freeze-drying. However,
`a complicating factor in the interpretation of the results is
`that both the ion product of water (pK ) and the dissocia-
`w
`tion constant pK of the imidazole moiety change with
`a
`temperature. In addition, when the water is frozen out, the
`concentration effect has also to be considered. The mean-
`
`Table 1
`Physical state of L-histidine after freeze-drying with and without a thermal
`a
`cycle from solutions of varying pH
`
`Freezing process
`
`pH 4
`
`pH 5
`
`pH 6
`
`pH 7
`
`pH 8
`
`No Thermal cycle
`Thermal cycle
`
`b
`A
`nt
`
`A
`nt
`
`A
`A
`
`c
`A
`c
`A
`
`c
`A
`C
`
`Fig. 5. Dependence of T during the second heating of frozen aqueous
`9
`g
`L-histidine and sucrose solutions on the weight to weight ratio of L-
`histidine or sucrose/salt.
`
`a
`
`b
`
`c
`
`A, X-ray amorphous; C, Crystalline; nt, not tested.
`The cake collapsed.
`Thin crystal clusters on the surface of the cakes.
`
`5
`
`

`

`306
`

`T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`ing of pH at low temperature is complex. The arbitrary
`o
`zero potential is the standard potential EH of the revers-
`ible hydrogen electrode, and this by definition is taken as
`zero at all temperatures. Accordingly it is not possible
`from the measurement from the hydrogen electrode po-
`tentials to compare exactly the true hydrogen activity at
`two different temperatures (Taylor, 1981). However, the
`ratio between the ionised and the un-ionised imidazole
`moiety in the freeze concentrated L-Histidine glass has to
`remain 1:1 since electroneutrality must be maintained. The
`carboxy and amino groups of L-histidine are fully ionised
`in the pH range investigated, so the chloride ions must be
`‘neutralised’ by the charged imidazole nitrogens. The
`dependence of the T of L-histidine on the pH has to be
`9
`g
`considered in the design of the freeze-drying cycle, since
`the T determines the maximum product temperature that
`9
`g
`can be used during primary drying. In practice it is usual to
`allow at
`least a 60.5 pH deviation from the specified
`value. For a formulation with a specified value of pH 7.0,
`these limits might give a 68C decrease in the T .
`9
`g
`
`3.4. Powder X-ray diffraction and water sorption/
`desorption
`
`The samples that were freeze-dried without a thermal
`cycle were subjected to powder X-ray diffraction analysis
`after a 2-year storage at 378C. The diffractograms (Fig. 6)
`
`showed that all the samples were X-ray amorphous. The
`samples freeze-dried with a thermal cycle (stored at 2–88C
`for 2 years) revealed some crystallinity, which decreased
`as the pH decreased (Fig. 7). A sample consisting of a
`mixture of sucrose and L-histidine (33%, w/w, of L-
`histidine and pH 7) is also shown in Fig. 7. This sample
`was X-ray amorphous, which indicates that the addition of
`sucrose inhibits crystallisation during freeze-drying and
`long-term storage. It is noteworthy that all the samples
`freeze-dried without a thermal cycle were X-ray amor-
`phous, which indicates that the amorphous to crystalline
`transformation of L-histidine is slow if the product
`is
`protected from moisture contact. The samples were ana-
`lysed in duplicate and the only difference between the
`diffractograms was the normal variation in amplitude
`which always occurs in powder X-ray diffraction. Fig. 8
`shows the water sorption and desorption profiles of
`samples freeze-dried at pH 5, 6 and 8, respectively. All the
`samples were highly hygroscopic and the sorption/ desorp-
`tion profiles indicate that
`they all start
`to crystallise
`between 20 and 40% RH. The end-point of the desorption
`profiles (weight ratio of water and L-histidine) indicated
`that the pH 5 sample crystallised as a monohydrate, the pH
`8 sample crystallised as an anhydrate and the pH 6 sample
`crystallised as a hemihydrate or a mixture of anhydrate and
`monohydrate. The duplicate samples showed very similar
`sorption/desorption profiles (not illustrated). The powder
`
`Fig. 6. Powder X-ray diffraction pattern of freeze-dried L-histidine (pH 4, 5, 6, 7 and 8) after a 2-year storage at 378C. Note that all samples were X-ray
`amorphous.
`
`6
`
`

`

`¨T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`307
`
`Fig. 7. Powder X-ray diffraction pattern of L-histidine (pH 6, 7 and 8) and L-histidine–sucrose (33%, w/ w), freeze-dried with a thermal cycle and after
`long-term storage under refrigeration. Note that the crystallinity is decreased at lower pH and that the sample with sucrose is completely X-ray amorphous.
`
`X-ray analysis of the freeze-dried samples after moisture
`exposure is shown in Fig. 9. The diffractograms showed
`that the solution freeze-dried at pH 5 crystallised as the
`L-histidine monohydrochloride monohydrate. The pH 8
`
`solution crystallised as L-histidine and the pH 6 sample
`crystallised as a 1:1 mixture of L-histidine monohydro-
`chloride monohydrate and L-histidine. The moisture-in-
`duced crystallisation was also readily observed in a
`polarising microscope by exposing the sample to moisture
`when crystallisation took place almost immediately.
`
`4. Conclusions
`
`L-Histidine may be regarded as a multifunctional protein
`stabiliser since it can function as a buffer and a metal ion
`scavenger and stabilise the protein in an amorphous phase.
`When formulating proteins with L-histidine, it is important
`to consider the pH value with regard to both possible
`crystallisation of L-histidine and its influence on the T .g
`9
`21
`21
`can be added to
`Metal
`ions such as Ca
`and Mg
`L-histidine without significant depression of T . The risk of
`9
`g
`crystallisation of amorphous L-histidine is low as long as
`the formulation is protected from moisture contact. The
`addition of sucrose reduces the risk of crystallisation and
`increases the T of the freeze-concentrated mixture.
`9
`g
`
`Acknowledgements
`
`Fig. 8. Water sorption/desorption of freeze-dried L-histidine (pH 5, 6 and
`8) as a function of the relative humidity. Note crystallisation between 20
`and 40% relative humidity.
`
`We gratefully acknowledge the assistance of Johan
`Andersson, Astra Arcus, who performed the moisture
`balance studies.
`
`7
`
`

`

`308
`

`T. Osterberg, T.Wadsten / European Journal of Pharmaceutical Sciences 8(1999)301–308
`
`Fig. 9. Powder X-ray diffraction pattern of freeze-dried L-histidine (pH 5, 6 and 8) after moisture-induced crystallisation. L-Histidine and L-histidine?HCl?
`H O (Sigma) were included as references.
`2
`
`References
`
`Arakawa, T., Prestrelski, S.J., Kenney, W.C., Carpenter, J.F., 1992.
`Factors affecting short-term and long-term stabilities of proteins. Adv.
`Drug. Deliv. Rev. 10, 1–28.
`Bush, L., Webb, C., Bartlett, L., Burnett, B., 1998. The formulation of
`recombinant factor IX: stability, robustness, and convenience. Semin.
`Hematol. 35, 18–21.
`Carpenter, J.F., Crowe, J.H., 1989. An infrared spectroscopic study of the
`interactions of carbohydrates with dried proteins. Biochemistry 28,
`3916–3922.
`Chang, B.S., Randall, C.S., 1992. Use of subambient thermal analysis to
`optimize protein lyophilization. Cryobiology 29, 632–656.
`Fox, T.G., 1950. Second-order transition temperatures and related prop-
`erties of polystyrene. I. Influence of molecular weight. J. Appl. Phys.
`21, 581–591.
`Franks, F., Hatley, R.H.M., Mathias, S.F.M., 1991. Materials science and
`the production of shelf stable biologicals. Pharm. Technol. Int. 3,
`24–34.
`Greenstein, J.P., Winitz, M, 1961. Metal and salt combinations with amino
`acids. In: Chemistry of the Amino Acids, Vol. 1, Wiley, New York, pp.
`560–681.
`Lamfrom, H., Nielsen, O., 1970. The iron catalysis of thioglycolate
`oxidation by oxygen. J. Am. Chem. Soc. 79, 1966–1970.
`
`MacKenzie, A.P., 1966. Basic principles of freeze-drying for pharma-
`ceuticals. Bull. Parenteral Drug Assoc. 20, 101–129.
`MacKenzie, A.P., 1977. The physico-chemical basis for the freeze-drying
`process. Dev. Biol. Stand. 36, 51–67.
`MacKenzie, A.P., 1985. Fundamentals and applications of freeze-drying
`to biological materials, drugs and foodstuffs. In: Proceedings of the
`Meeting of the Commission C1, Tokyo, Japan, Refrigeration Science
`and Technology, pp. 21–34.
`¨Osterberg, T., Fatouros, A., Mikaelsson, M., 1997. Development of
`freeze-dried formulation of recombinant Factor VIII SQ. Pharm. Res.
`14, 892–898.
`Pikal, M.J., 1990. Freeze-drying of proteins. Part II: Formulation selec-
`tion. BioPharm 3, 26–30.

`Shihong, L., Schoneich, C., Wilson, G.S., Borchardt, R.T., 1993. Chemi-
`cal pathways of peptide degradation. V. Ascorbic acid promotes rather
`than inhibits the oxidation of methionine to methionine sulfoxide in
`small model peptides. Pharm. Res. 10, 1572–1579.
`Sundberg, R.J., Martin, B.R., 1974. Interactions of histidine and other
`imidazole derivatives with transition metal
`ions in chemical and
`biological systems. Chem. Rev. 74, 471–517.
`Taylor, M.J., 1981. The meaning of pH at low temperatures. Cryo-letters
`2, 231–239.
`
`8
`
`

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