throbber
Pharmaceutical Research, Vol. 14, No. 8, 1997
`
`Review Article
`
`Rational Design of Stable Lyophilized Protein Formulations:
`Some Practical Advice
`
`John F. Carpenter,1•2•6 Michael J. Pikal,3 Byeong S. Chang,4 and Theodore W. Randolph1•5
`
`Received March 7, 1997; accepted May 15, 1997
`
`KEY WORDS: protein drugs; design of fonnulations; lyophilization; stabilization of proteins.
`
`WHY USE LYOPHILIZATION TO PREPARE
`STABLE PROTEIN DRUG PRODUCTS?
`
`Early in the development of a protein therapeutic 1t 1s
`essential to design a formulation that is stable during shipping
`and long-term storage. Obviously, an aqueous liquid formula(cid:173)
`tion is the easiest and most economical to handle during manu(cid:173)
`facturing, and is the most convenient for the end user. However,
`many proteins are susceptible to chemical (e.g., deamidation
`or oxidation) and/or physical degradation (e.g., aggregation and
`precipitation) in liquid formulations ( 1,2). It may be possible
`to design an aqueous formulation to slow protein degradation
`adequately, under controlled storage conditions (i.e., constant
`temperature and minimal agitation). However, during shipping,
`when precise control of conditions is not always feasible, prod(cid:173)
`ucts can be subjected to numerous stresses that denature pro(cid:173)
`teins. These include agitation, high and low temperatures, and
`freezing (2). Furthermore, although a formulation and shipping
`system might be designed to circumvent damage from these
`stresses, it still may not be possible to inhibit damage suffi(cid:173)
`ciently during long-term storage. For example, there are cases
`where conditions that minimize chemical degradation foster
`physical damage and vice versa (1,2). Then, conditions that
`provide a compromise affording the requisite long-term stability
`cannot be found.
`All of these difficulties theoretically can be avoided with
`a properly prepared lyophilized formulation. In the dried solid,
`degradative reactions can be avoided or slowed sufficiently,
`such that the protein product remains stable for months or
`years at ambient temperatures (3-6). Furthermore, short-term
`
`1 University of Colorado Center for Phannaceutical Biotechnology,
`School of Phannacy, University of Colorado Health Sciences Center,
`Denver, Colorado 80262.
`2 Department of Pharmaceutical Sciences, School of Phannacy, Univer(cid:173)
`sity of Colorado Health Sciences Center, Denver, Colorado 80262.
`3 School of Phannacy, University of Connecticut, Storrs, Connecti(cid:173)
`cut 06269.
`4 Amgen, Inc., Thousand Oaks, California.
`5 Department of Chemical Engineering, University of Colorado, Boul(cid:173)
`der, Colorado.
`6 To whom correspondence should be addressed. (e-mail: John.
`Carpenter@UCHSC.edu)
`
`excursions in temperature control during shipping are usually
`not damaging to a lyophilized protein (6). Even in cases where
`two, or more, degradative pathways require different conditions
`for maximum thermodynamic stabilization, the reduced reaction
`rates in a dried product can allow for long-term stability. Thus,
`in general, whenever preformulation studies indicate that suffi(cid:173)
`cient protein stability cannot be achieved in aqueous liquid
`formulations,
`lyophilization provides
`the most attractive
`alternative.
`However, lyophilization requires sophisticated processing
`and is time consuming and expensive, relative to simply filling
`vials with a liquid formulation (3-10). Also---0f greatest con(cid:173)
`cern for the current review-without appropriate stabilizing
`excipient(s) most protein preparations are at least partially dena(cid:173)
`tured by the freezing and dehydration stresses encountered dur(cid:173)
`ing lyophilization (2,3-6, 11-16). The result is often irreversible
`aggregation of a fraction of the protein population, either imme(cid:173)
`diately after processing or after storage (e.g., 15,16). Because
`most protein drugs are delivered parenterally, only a few percent
`of aggregated protein will be unacceptable. Finally, simply
`designing a formulation that allows the protein to survive the
`lyophilization process does not assure stability during long(cid:173)
`term storage in the dried solid (6,13-16). A poorly formulated
`lyophilized product, in which the protein is sufficiently reactive
`to require storage at subzero temperature, should not be consid(cid:173)
`ered a success.
`The purpose of this mini-review is to provide some practi(cid:173)
`cal guidelines for designing formulations that protect proteins
`during freezing and drying, and that are stable during shipping
`and long-term storage at ambient temperatures. Also, as will
`be discussed briefly, formulations must be designed with consid(cid:173)
`eration of the physical constraints on processing conditions
`needed to obtain a proper final cake with a low residual mois(cid:173)
`ture. All of these issues have been reviewed previously in detail
`(3-10). Furthermore, relevant new reviews by us and others
`will appear this year in books edited by Vincent Lee and Louis
`Rey. We will not discuss the design and optimization of lyophili(cid:173)
`zation cycles. Nor will we digress from practical advice about
`excipient choices to address the debates about the mechanisms
`by which these compounds stabilize proteins (see 2,5,6, 11-16).
`The pharmaceutical scientist who has had extensive experience
`bringing lyophilized protein products to market may not benefit
`
`969
`
`0724-8741/97/0800-0969$12.50/0 © 1997 Plenum Publishing Corporation
`
`AMNEAL EX. 1011
`
`

`

`970
`
`Carpenter, Pikal, Chang, and Randolph
`
`greatly from this review. Rather, our goal is to provide a starting
`point for the researcher for whom design of stable lyophilized
`protein formulations is still a new and major challenge.
`
`WHAT CONSTRAINTS GOVERN THE DESIGN OF
`THE FORMULATION
`
`There are so many factors to consider when designing a
`proper lyophilized formulation, that the task when viewed as
`a whole can appear overwhelming. This need not be the case,
`if the major constraints governing success are well understood.
`
`Protein Stability
`
`First, it must be remembered that the whole reason for
`lyophilizing the product is because the protein of choice is
`unstable. The most sensitive element in the formulation is the
`protein, and the primary concern in formulation design must
`be the choice of excipients that provide optimal stability. This
`is the issue on which we will focus in detail below.
`
`Final Product Configuration
`
`Secondly, the final product configuration must be clearly
`defined prior to starting formulation efforts. Issues to be consid(cid:173)
`ered include route of administration, which is often parenteral,
`other agents to be co-administered to the patient, product vol(cid:173)
`ume, protein concentration, and whether the product can be
`lyophilized in vials or whether alternative systems such as
`syringes must be employed. Also, if the final product is intended
`for multi-use, it will necessary to include a preservative in the
`formulation, which may reduced protein stability.
`
`Formulation Tonicity
`
`In choosing excipients, designing an isotonic solution
`might be a concern. Mannitol or glycine are usually good
`choices as tonicity modifiers. As explained below, these excipi(cid:173)
`ents are often preferable to NaCl, which due to its relatively
`low eutectic melting and glass transition temperatures, can make
`a formulation more difficult to lyophilize properly (3-10, 17-20).
`Also, if the product has a relatively low mass of protein per
`vial, often it will necessary to have a bulking agent in the
`formulation to prevent the protein from being lost from the vial
`during drying (e.g., 4-6). Mannitol or glycine can also serve
`this role because they usually crystallize to a substantial degree
`during lyophilization and form a mechanically strong cake
`(4-6). However, it must be realized that crystalline excipients
`when used alone will usually not provide adequate stability to
`most proteins during processing or storage in the dried solid
`(12-14).
`
`Cake Structure
`
`Finally, the dried product must have an elegant cake struc(cid:173)
`ture, which is mechanically strong and has not undergone any
`collapse and/or eutectic melting and in which the residual mois(cid:173)
`ture is relatively low (ca. 1 g H2O/100 g dried solid). If the
`product collapses, it will not only be aesthetically unacceptable,
`but also it could have excessively high residual moisture, and
`reconstitution time will be prolonged (3-6,17,18).
`
`Product Glass Transition Temperature
`
`Also, to assure long-term stability of the protein in the
`dried solid, the glass transition temperature (Tg) of the amor(cid:173)
`phous phase in the product, which contains the protein, must
`exceed the planned storage temperature ( 4-6, 14-16). Since
`water is a plasticizer of the amorphous phase (3-6,9, 17, 18),
`low residual moisture is needed to insure that Tg is greater
`than the highest temperature encountered during shipping and
`storage (usually greater than 40°C).
`
`Product Collapse Temperature
`
`In general, achieving these goals requires maintaining the
`product temperature below its glass transition temperature dur(cid:173)
`ing the lyophilization cycle (3-10, 17, 18). During primary dry(cid:173)
`ing, when ice is sublimed, the product must be maintained
`below the collapse temperature, which usually coincides with
`the thermotropic transition that has been referred to the glass
`transition temperature of the maximally freeze-concentrated
`amorphous phase of the sample (Tg') or as the softening temper(cid:173)
`ature of the amorphous phase (Ts) (3-10,17,18). Also, it is
`necessary to keep the product temperature below the eutectic
`melting temperature of any crystalline component. In practice,
`these temperatures can be determined using either differential
`scanning calorimetry (DSC) or freeze-drying microscopy. The
`ability to determine collapse temperature is essential to formula(cid:173)
`tion development (19 ,20).
`Drying a product below the collapse temperature carries
`a price (3-10). The lower the sample temperature, the slower
`and more expensive the drying cycle becomes. In general,
`freeze-drying below -40°C is not practical (3-10). Also, there
`are physical limits in the temperatures to which samples can
`be reduced, which are dependent on the lyophilizer and sample
`configuration (3-10). As the formulation is being developed, the
`pharmaceutical scientist should work closely with the process
`engineers, who will be designing the lyophilization cycles. It
`is especially important to know how the large-scale lyophilizers,
`which will be used for commercial production, compare to the
`research-scale unit that is used during formulation development.
`Often, the large units do not have the same level of control of
`process parameters as do the small research units, and in part
`due to the large size of a production unit, intervial variation in
`product temperature during the process may be greater. Finally,
`input from a researcher knowledgeable in the physics of freeze(cid:173)
`drying will help prevent the formulation scientist from arbi(cid:173)
`trarily rejecting useful formulations. There are ways (see
`reviews 3-10) in which the process parameters can be manipu(cid:173)
`lated such that relatively rapid and controlled drying can be
`achieved with products that have relatively low collapse
`temperatures.
`It is clear that one goal of formulation design is to provide
`the highest collapse temperature that is practical, within the
`constraints of maintaining protein stability. The collapse tem(cid:173)
`perature (i.e., the Tg') of the product will be dictated primarily
`by the formulation composition. If the protein is present at a
`level exceeding about 20% (wt/wt) of all solute it can have a
`relatively large effect on Tg'. Although it is often difficult to
`measure the Tg' of pure protein solutions with DSC, it has
`been found that adding increasing amounts of protein to most
`formulations leads to a higher Tg' (S.D. Allison, B.S. Chang,
`T.W. Randolph, M.J. Pikal and J.F. Carpenter, unpublished
`
`AMNEAL EX. 1011
`
`

`

`Design of Stable Lyophilized Protein Formulations
`
`971
`
`observations). By extrapolation it appears that pure protein
`solutions have a Tg' of about - l0°C, which is much higher
`than that of most pure excipient solutions (e.g., Tg' of sucrose
`is about - 32°C). Thus, from a process economy viewpoint,
`one desires a high ratio of protein to stabilizer in the formulation
`(cf. 6). However, stability normally increases as the weight
`ratio of stabilizer to protein increases, so typically a compromise
`must be made between providing a high collapse temperature
`and adequate protein stabilization ( e.g., 2,4-6, 11, 16). Also, as
`will be discussed below, protein resistance to freezing damage
`often improves as the protein concentration increases (2,6,22).
`Thus, in general stability is best at both high protein concentra(cid:173)
`tion and high weight ratio of stabilizer to protein. Therefore,
`in tum, stability optimization may lead to very high total solids
`content, which creates processing difficulties. Formulations
`with total solids in excess of 10% (w/w) may be difficult to
`process (3,7-10).
`Also, the manner in which the formulation is treated prior
`to applying a vacuum can alter the Tg'. Usually such treatment
`involves an annealing step, which results in removing some
`fraction of a given component from the amorphous phase (6).
`For example, if glycine is used as a crystalline bulking agent,
`depending on the freezing protocol, a significant fraction of
`the glycine molecules may remain in the amorphous phase of
`the sample (6). Glycine has a relatively low Tg' (e.g., ca.
`-42°C; 6,20). Thus, it is important to crystallize as much as
`possible, which in turn should increase the Tg' of the amorphous
`phase and make drying more rapid and economical. To design
`the optimum protocol for excipient crystallization, DSC can be
`used to simulate the processing conditions used during freezing
`and annealing. This approach is described in Carpenter and
`Chang (6).
`
`AT WHAT STEPS IS STABILIZATION OF THE
`PROTEIN REQUIRED?
`
`Essentially every step from vial filling to final reconstitu(cid:173)
`tion of the dried product can damage the protein and require
`formulation components to inhibit degradation ( 1,2,6, 11,21,22).
`During the rapid steps (e.g., filling, freezing, drying and rehy(cid:173)
`dration) the major problem is usually physical damage, which
`is typically manifested as formation of oligomeric and/or precip(cid:173)
`itated protein molecules (1,2,6,15,21,22). Normally the transi(cid:173)
`tion from solution to solid slows the rate of physical changes
`more than it slows chemical changes, so chemical degradation
`in the dried solid is often the more serious storage stability
`problem (e.g., 6,15,16). However, protein aggregates can form
`during storage/reconstitution (e.g., 6, 13-16). These degradative
`processes can be minimized if protein unfolding (here, meaning
`even a small fraction of the total molecular population) is
`inhibited during the most damaging stresses of freezing and
`drying (6,15,16). Thus, a primary focus of formulation design
`should be protecting the protein during these steps, so that the
`dried formulation immobilizes the native protein in a chemically
`inert solid matrix having both high Tg and low residual mois(cid:173)
`ture (5,6,14,16).
`
`Stabilization During Freezing
`
`stabilizers (see below) in the formulation. In general, the three
`most important parameters to consider are protein concentra(cid:173)
`tion, buffer choice, and freezing protocol (2,6,21-24).
`Increasing protein concentration leads to increased resis(cid:173)
`tance to denaturation during freezing (2,6,22,24). This phenom(cid:173)
`enon can be demonstrated by simply determining the percentage
`protein aggregated after freeze-thawing, which varies inversely
`with protein concentration (e.g., 22). Normally, it would be
`expected that increasing protein concentration would increase
`aggregation, and this would be the case if the fraction of protein
`molecules unfolded during freezing were independent of con(cid:173)
`centration. However, it is now thought that increasing protein
`concentration directly
`reduces
`freezing-induced protein
`unfolding. It has been speculated that damage during freezing
`involves protein denaturation during formation of the ice-water
`interface (21,22). Assuming that only a finite number of protein
`molecules can be denatured at this interface, then increasing
`the initial protein concentration will lead to a smaller percentage
`of damaged molecules. For practical purposes, it is sufficient
`simply to consider protein concentration as an important vari(cid:173)
`able to examine, and to include the highest possible concentra(cid:173)
`tion in testing during formulation development.
`Buffer choice can also be critical. The main culprits here
`are sodium phosphate and potassium phosphate, which can
`undergo drastic changes in pH during freezing and annealing
`(6,23,24). With sodium phosphate, the dibasic form will readily
`crystallize, resulting in a frozen sample in which the pH in the
`remaining amorphous phase (containing the protein) can be
`reduced to 4 or lower (23,24). With potassium phosphate, the
`dihydrogen salt crystallizes, giving a final pH near 9 (23,24).
`The risk of alteration in pH and its damage to proteins can be
`minimized by increasing the initial cooling rate, limiting the
`duration of annealing steps and minimizing the buffer concen(cid:173)
`tration, all of which reduce opportunity for salt crystallization
`(6,24). Rapid freezing, without annealing also limits the length
`of exposure of protein to denaturing conditions in the frozen
`state 6,24). Although other excipients can aid in inhibiting the
`pH change (24), the best approach is to avoid using sodium
`phosphate or potassium phosphate buffers. Buffers that have
`minimal pH change upon freezing include citrate, histidine and
`Tris (22,24; T.J. Anchordoquy and J.F. Carpenter, unpub(cid:173)
`lished observations).
`In studies in which complications due to buffer pH changes
`have been avoided, it has been found that the degree of protein
`damage during freezing correlates directly with cooling rate,
`with more damage found at higher cooling rates where surface
`area of ice is larger (21,22). It has been speculated that this is
`due to protein denaturation during the formation of the ice(cid:173)
`water interface (21,22). More rapid cooling leads to smaller
`ice crystals, which have a greater surface area to volume ratio
`than larger crystals. Since cooling rates will usually be dictated
`by the physical constraints of the Iyophilizer, excessively rapid
`cooling probably will not be a problem (3-10). However, some
`proteins are so sensitive to freezing, that even with slow, con(cid:173)
`trolled cooling they will be denatured (e.g., 21,24).
`
`Stabilization During Drying and Storage in the Dried
`Solid
`
`Whether a given protein is susceptible to freezing damage
`depends of many factors, beyond the inclusion of the appropriate
`
`Even if the entire population of protein molecules survives
`the freezing step, there will be denaturation during subsequent
`
`AMNEAL EX. 1011
`
`

`

`972
`
`Carpenter, Pikal, Chang, and Randolph
`
`dehydration, unless the appropriate stabilizers are added
`(15, 16,25-28). Simply stated, removal of the protein molecule's
`hydration shell, which occurs during lyophilization, destabilizes
`the native conformation (15, 16,25-28). To date, infrared spec(cid:173)
`troscopic studies with dozens of proteins have shown that, in
`the absence of the appropriate stabilizer(s) (e.g., sucrose) pro(cid:173)
`teins will be unfolded in the dried solid (15,16,25-28). If sam(cid:173)
`ples are rehydrated immediately, the degree of damage (e.g.,
`percent of aggregation) correlates directly with how "non(cid:173)
`native" the infrared spectrum of the dried protein appeared
`(15,16,25-28). Thus, reducing post-rehydration damage is
`dependent on minimizing the unfolding during freezing and
`drying. Moreover, even if 100% native molecules are recovered
`in samples rehydrated immediately, there can be a substantial
`fraction ofunfolded molecules in the dried solid (15,16,25-28).
`Intramolecular refolding during rehydration can dominate the
`intermolecular interactions leading to aggregation, thereby giv(cid:173)
`ing 100% native protein on reconstitution.
`Fortunately, appropriate excipients can prevent or at least
`minimize unfolding, and the success of the formulation can
`be judged immediately by examining the protein secondary
`structure
`in
`the dried solid with
`infrared spectroscopy
`(15,16,25-28). More importantly, in the few studies published
`to date, it has been shown that stability during long-term storage
`in the dried solid is dependent of retention of native protein
`during freeze-drying (15, 16). Even for samples stored at temper(cid:173)
`atures well below the formulation Tg, damage arose rapidly
`(e.g., within weeks) if the protein was unfolded in the dried
`solid. Therefore, infrared spectroscopy, which can be used
`immediately after lyophilization
`to determine
`if protein
`unfolded has arisen, should be considered another essential tool
`for the protein formulation scientist.
`
`WHICH EXCIPIENTS ARE THE BEST FIRST
`CHOICES?
`
`After this review of all of the dangers of lyophilization
`and all the factors to be considered it might seem that rapid
`development of a stable lyophilized formulation would be an
`impossible task. Fortunately with a rational approach to formu(cid:173)
`lation design, most formulation problems are quickly resolved.
`Here we will provide the rationale for the initial choices of
`formulation components. In some cases, the "initial formula(cid:173)
`tion" may be all that is needed for the final marketed product.
`The composition to be given, with various minor modifications,
`has already been used with success with protein drugs (e.g.,
`16). We wish to stress that for any lyophilized formulation, the
`minimum number of components necessary for protein stability
`and cake structure should be used. No excipient should be
`added unless there are data to document that it has a beneficial
`role in the formulation.
`
`Specific Conditions for Stability of a Given Protein
`
`Before choosing the appropriate "general" stabilizers,
`which are effective at protecting most proteins, it is absolutely
`essential that the formulation be optimized for the specific
`factors that increase the physical and chemical stability of a
`given protein. For example, simply avoiding extremes in pH
`can drastically reduce the rate of deamidation (1). Moreover,
`it has been found that the resistance of a protein to unfolding
`
`during freeze-drying can be dramatically increased by optimiz(cid:173)
`ing the pH of solution (e.g., 15). Also, other specific ligands
`that increase protein stability (e.g., by increasing the free energy
`of unfolding) should be investigated. The stabilizing effects of
`heparin and other polyanions on growth factors (e.g., 29) pro(cid:173)
`vide a good example. Another important factor to be considered
`is the effect of ionic strength on protein unfolding and aggrega(cid:173)
`tion. It must be recognized that during freezing, the ionic
`strength may increase SO-fold as ice formation concentrates
`all solutes (5,6,9,19). The persons responsible for bulk drug
`purification and pharmaceutical preformulation often already
`have insight into these issues. Thus, it is imperative that the
`these people, prior to
`formulation scientist confer with
`embarking of design of a lyophilized formulation.
`Even with specific solution conditions optimized for pro(cid:173)
`tein stability, it probably will be necessary to add other protec(cid:173)
`tive excipients, if the protein is to survive lyophilization and
`long-term storage in the dried solid. First, let us consider some
`compounds that have been used for lyophilized protein formula(cid:173)
`tion but which do not provide stability and may actually foster
`damage during storage. We will then provide an outline of a
`simple, but effective formulation, and the rationale for the
`choice of the components will be discussed.
`
`Excipients that Can Fail to Stabilize Proteins
`
`With the goal of obtaining a strong cake structure during
`a rapid lyophilization cycle, polymers such as dextran and
`hydroxyethyl starch, which have relatively high collapse tem(cid:173)
`peratures, are attractive excipients. Also, the Tg of the final
`dried product will be high (e.g., >90°C) with these polymers
`( 15). Unfortunately, these polymers do not inhibit protein
`unfolding during lyophilization and they typically fail to provide
`stability during subsequent storage (15,30). The failure to inhibit
`lyophilization-induced denaturation is presumably because the
`polymers are too bulky to hydrogen bond to the protein in the
`place of the water that is lost during dehydration and/or because
`the polymers form a separate amorphous phase from the protein
`(5,6). Although when used alone such polymers are not good
`choices as stabilizers, as described below, they could be prove
`useful
`in combination with certain disaccharide protein
`stabilizers.
`Among the numerous compounds tested it appears that
`the most effective stabilizers during the lyophilization cycle
`are disaccharides (2,5,6,11,15,16,25-28). However, one group
`of compounds that should be avoided are the reducing sugars.
`These compounds may effectively inhibit protein unfolding
`during the lyophilization cycle, but during storage in the dried
`solid they have the propensity to degrade proteins via the Mail(cid:173)
`lard reaction between carbonyls of the sugar and free amino
`groups on the protein (31). The result can be a brown syrup
`containing degraded protein instead of a white cake containing
`active protein drug. Usually, the only way to slow this process
`significantly is to store the product at subzero temperatures,
`which defeats the purpose of a lyophilized product. Compounds
`in this undesirable category include glucose, lactose, maltose
`and maltodextrins.
`As noted earlier, crystalline bulking agents such as manni(cid:173)
`tol and glycine do not provide protection during lyophilization
`(6, 11, 12,25). However, some effective lyophilized formulations
`employing mixtures of these two agents have been developed
`
`AMNEAL EX. 1011
`
`

`

`Design of Stable Lyophilized Protein Formulations
`
`973
`
`and marketed. In these cases the appropriate ratio of manni(cid:173)
`tol:glycine led to a significant fraction of the compounds
`remaining amorphous (e.g., 30). Presumably this amorphous
`fraction was sufficient to inhibit protein unfolding during lyoph(cid:173)
`ilization and to provide stability during long-term storage. How(cid:173)
`ever, we caution against such an approach because achieving
`just the right processing conditions, in combination with the
`appropriate excipient ratio, can be time consuming and tricky.
`
`Rational Choice of Stabilizing Excipients
`
`So what are appropriate, rational choices for excipients?
`To provide a concrete example, let's make the following
`assumptions about a fictitious case. 1) The protein drug will
`be formulated at 2 mg/ml. 2) The major routes of degradation are
`aggregation immediately after lyophilization/rehydration and
`deamidation during storage in the dried solid. 3) Optimizing
`specific conditions (e.g., using a citrate buffer at pH 6.0) only
`reduces aggregation upon freeze-drying and reconstitution to
`about 10% and deamidation still proceeds at an unacceptably
`rapid rate during storage, even when the product is stored 20°C
`below its Tg. 4) A crystalline bulking agent (e.g., mannitol) is
`desired to form a mechanically strong and elegant cake.
`At this point, the major component missing is a nonreduc(cid:173)
`ing disaccharide, which forms an amorphous phase with the
`protein in the dried solid and serves as the primary stabilizer.
`The main choices are sucrose or trehalose (5,6). These com(cid:173)
`pounds are relatively effective at protecting proteins during
`freezing and usually excellent at inhibiting unfolding during
`dehydration (5,6,15,16,25-28). Freezing protection depends on
`the initial bulk concentration of the sugar, and sometimes con(cid:173)
`centrations exceeding 5% (wt/vol) are needed to maximize
`stabilization (2,6). In contrast, protection during drying depends
`on the final mass ratio between the sugar and the protein (5,6).
`Generally, a weight ratio of sugar to protein of at least I: 1 is
`required for good stability, with optimal stability being reached
`at around 5: 1. In practice, it is not necessary to determine the
`most appropriate concentration for each type of protection.
`Rather, with the protein concentration held constant, a range
`of sugar concentrations can be tested during formulation screen(cid:173)
`ing to discern the optimal concentration for retention of native
`protein in the dried solid and the resultant reduction in aggrega(cid:173)
`tion upon rehydration.
`In general, the optimal sugar concentration for stabilizing
`the protein during lyophilization will also provide storage stabil(cid:173)
`ity, if the final dried powder has a Tg well above the storage
`temperature (5,6, 15, 16). For example, assuming that a high
`room temperature of about 30°C is the maximum intended
`storage temperature, then a product containing a native protein
`and with a Tg of > 50°C should be stable. Since residual
`moisture lowers Tg, the condition Tg > 50°C must apply to
`the maximum water content allowed by the product specifica(cid:173)
`tions. It is imperative that DSC be used to measure the Tg of
`each product to be certain that this goal is achieved.
`Both sucrose and trehalose have advantages and disadvan(cid:173)
`tages. 1) Trehalose has a higher Tg at any moisture content
`and, thus, is more easily lyophilized (32). In addition, the condi(cid:173)
`tion Tg > 50°C will hold at higher residual water contents for
`trehalose. However, a skilled process engineer should be able
`to design economical, effective cycles for either sugar. Also,
`in products with a relatively high protein concentration, the
`
`protein could contribute to an increased Tg, which serves to
`minimize the advantages of trehalose. 2) Trehalose is also more
`resistant than sucrose to acid hydrolysis. Hydrolysis of these
`disaccharides produces reducing sugars, which must be avoided.
`Usually this is not a problem, unless very low pH's of around
`4 or lower are employed. 3) Sucrose appears to be more effective
`at inhibiting unfolding during lyophilization (unpublished
`observations). This difference has been most obvious when
`there is a relatively high protein concentration and a need to
`employ a relatively high initial concentration of sugar. This
`need can develop when the protein is very unstable during
`freezing and/or there is a relatively high protein content in
`the dried solid. Evidence to date indicates that less effective
`stabilization by trehalose is due to the greater propensity of
`this sugar to phase separate from polymers (e.g., the protein in
`a formulation) during freezing and drying (S.D. Allison, T.W.
`Randolph, B.S. Chang and J.F. Carpenter, unpublished observa(cid:173)
`tion). Whether this is a problem with a given formulation cannot
`be predicted. Hence, the capacity to protect a protein must be
`examined for each formulation. 4) Sucrose is commonly used
`in parenteral products that are approved by the Food and Drug
`Administration (33). In contrast, trehalose has not yet been
`used in an approved product, probably because to date there
`has not been sufficient practical bendit to justify using this
`sugar in place of sucrose. Safety of t:rehalose will most likely
`not be a concern. Thus, if there is a clear advantage of trehalose
`over sucrose in a given product, use of trehalose should not
`hinder regulatory approval.
`At this point, our example formulation might be complete,
`as is the case for many proteins. However, let's assume that
`even with sucrose completely inhibiting detectable protein
`unfolding, as assessed with structural analysis of the dried solid
`with infrared spectroscopy, after rehydration there is still about
`1 % aggregated protein. Since there are no aggregates in the
`starting material, it is assumed that at some point during freezing
`and drying a very small fraction of the protein population was
`unfolded. During rehydration some of these molecules refolded,
`but others formed aggregates. This actually appears to be a
`common problem, as is the format

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