throbber
DRUGS AND THE PHARMACEUTICAL SCIENCES
`
`VOLUME 175
`
`
`
`
`
`MeUME)
`TT
`NAT aTTT
`
`MYLAN - EXHIBIT 1020
`
`edited by
`Eugene J. McNally
`Jayne E. Hastedt
`
`informa
`healthcare
`
`MYLAN - EXHIBIT 1020
`
`

`

`Protein Formulation
`and Delivery
`Second Edition
`
`Edited by
`Eugene J. McNally
`Gala Biotech, a Catalent Pharma Solutions Company
`Middleton, Wisconsin, USA
`Jayne E. Hastedt
`ALZA Corporation
`Mountain View, California, USA
`
`McNally_978-0849379499_TP.indd 2
`
`9/26/07 11:10:41 AM
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Copyright_page
`
`Informa Healthcare USA, Inc.
`52 Vanderbilt Avenue
`New York, NY 10017
`
`© 2008 by Informa Healthcare USA, Inc.
`Informa Healthcare is an Informa business
`
`No claim to original U.S. Government works
`Printed in the United States of America on acid-free paper
`10 9 8 7 6 5 4 3 2 1
`
`International Standard Book Number-10: 0-8493-7949-0 (Hardcover)
`International Standard Book Number-13: 978-0-8493-7949-9 (Hardcover)
`
`This book contains information obtained from authentic and highly regarded sources. Reprinted material is
`quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts
`have been made to publish reliable data and information, but the author and the publisher cannot assume
`responsibility for the validity of all materials or for the consequence of their use.
`
`No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic, mechan-
`ical, or other means, now known or hereafter invented, including photocopying, microfi lming, and recording, or
`in any information storage or retrieval system, without written permission from the publishers.
`
`For permission to photocopy or use material electronically from this work, please access www.copyright.
`com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood
`Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profi t organization that provides licenses and
`registration for a variety of users. For organizations that have been granted a photocopy license by the CCC,
`a separate system of payment has been arranged.
`
`Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used
`only for identifi cation and explanation without intent to infringe.
`
`Library of Congress Cataloging-in-Publication Data
`
`Protein formulation and delivery / edited by Eugene J. McNally, Jayne E. Hastedt. -- 2nd ed.
`
`
` p. ; cm. --
`(Drugs and the pharmaceutical sciences ; 175)
`
`Includes bibliographical references and index.
`
`ISBN-13: 978-0-8493-7949-9 (hardcover : alk. paper)
`
`ISBN-10: 0-8493-7949-0 (hardcover : alk. paper)
` 1. Protein drugs--Dosage forms.
`I. McNally, Eugene J., 1961- II. Hastedt, Jayne E.
`III. Series: Drugs and the pharmaceutical sciences ; v.175.
`
`[DNLM: 1. Protein Conformation. 2. Drug Delivery Systems. 3. Drug Design.
`4. Drug Stability. 5. Proteins--administration & dosage. W1 DR893B v.175 2007
`/ QU 55.9 P9667 2007]
`
`RS431.P75P77 2007
`615’.19--dc22
`
`Visit the Informa Web site at
`www.informa.com
`
`and the Informa Healthcare Web site at
`www.informahealthcare.com
`
`2007023435
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`9
`Rational Choice of Excipients for Use
`in Lyophilized Formulations
`
`Evgenyi Y. Shalaev
`Parenteral Center of Emphasis, Groton Laboratories, Pfi zer Inc.,
`Groton, Connecticut, U.S.A.
`
`Wei Wang
`Pharmaceutical Sciences—Global Biologics, Pfi zer Inc.,
`Chesterfi eld, Missouri, U.S.A.
`
`Larry A. Gatlin
`Parenteral Center of Emphasis, Groton Laboratories, Pfi zer Inc.,
`Groton, Connecticut, U.S.A.
`
`INTRODUCTION
`
`The majority of protein drugs are delivered by the injection route, although there
`is an increasing interest in alternative delivery routes, e.g., pulmonary. Ready-to-
`use liquid formulations are preferred injectable dosage forms because they are
`considered easier to manufacture and administer. However, the majority of pro-
`teins are not suffi ciently stable in aqueous media to provide adequate commercial
`shelf-life and this limits the development of protein pharmaceuticals as ready-
`to-use injectables. Freeze-drying is an established process to increase long-term
`stability of proteins and achieve an acceptable shelf-life (1). In some cases, as
`with proteins intended for administration by inhalation, spray-drying is used (2).
`It is also possible to simply dry protein solutions slowly at ambient temperatures
`under vacuum (3). This chapter deals with freeze-dried protein formulations as
`they are the most common commercial dosage forms. However, general prin-
`ciples can be applied to other dehydration processes such as spray-drying and
`vacuum-drying.
`
`197
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`198
`
`Shalaev et al.
`
`Essentially all protein formulations contain one or more inactive ingredi-
`ents (excipients). Excipients are used to facilitate the formulation manufactur-
`ing process, ensure stability of the active ingredient during processing, storage,
`and administration, minimize adverse effects upon administration (e.g., mini-
`mize pain upon injection), and ensure desirable bioavailability and (for sustained
`release dosage forms) release profi les. Each excipient in the formulation requires
`justifi cation for its use and an appropriate rationale for the level selected. Only
`excipients that are essential for performance and/or stability of a dosage form and
`suitable for injectable products are allowed to be included.
`The majority of lyophile protein dosage forms contain buffer and a bulk-
`ing agent, the latter often playing a dual role for both pharmaceutical elegancy
`and cryo- and lyoprotection, to achieve stability during processing and the shelf-
`life. In addition, many protein formulations contain additional stabilizers, e.g., a
`surfactant, and occasionally an antioxidant or a chelating agent. In some cases,
`a tonicity modifi er, a solubilizer, a processing aid, or an antimicrobial agent may
`be used. It is notable that the active ingredient level in the formulation can range
`from as high as close to 100% to as low as a few parts per million. Therefore, it
`is also possible to have a large range of excipient levels in the fi nal formulation.
`It should be mentioned also that excipients, which are important for the recon-
`stituted solution, e.g., antimicrobial agents or tonicity modifi ers, can be added
`with the diluent rather than being incorporated into the lyophile cake. Generally,
`selection of a proper excipient should take into account (i) the type of product,
`(ii) the delivery route, dose, and administration frequency, (iii) the chemical and
`physical properties of the excipient, (iv) potential interactions with other product
`components, and (v) the container/closure system.
`It is typically advantageous to choose formulation excipients that will not
`only enable the product to meet its critical quality parameters but also facili-
`tate the freeze-drying process because of the high cost/long processing times
`for this unit operation. This is especially critical when developing formulations
`for unique package systems such as dual chamber syringes, because of the dif-
`fi culties encountered in uniform drying in these packages. Therefore, selection of
`excipients that can potentially increase the collapse or eutectic temperatures of the
`frozen solution can greatly facilitate the drying process, thereby reducing cost and
`processing times. It is also important to select excipients whose vapor pressure is
`suffi ciently low so as not to permit its removal during the lyophilization process.
`There are a number of reviews available on different aspects of protein
`freeze-drying (1,4–10). In particular, it has been recognized that understand-
`ing phase behavior is a key for lyophile formulation and process development.
`Therefore, we start with a discussion of phase behavior of excipients during
`manufacturing and storage. Phase transitions have a major impact on stability
`and performance of protein dosage forms. For example, crystallization of a lyo-
`protector may result in protein destabilization during freeze-drying and storage.
`Description of excipients based on their functional role in protein formulations is
`given in the section titled Role and Properties of Excipients, followed by practical
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`Rational Choice of Excipients for Use in Lyophilized Formulations
`
`199
`
`advice on rationale excipient choice (based on both functional and physical chem-
`ical properties of excipients).
`
`PHASE BEHAVIOR OF EXCIPIENTS DURING LYOPHILIZATION
`AND STORAGE: GENERAL CONSIDERATIONS
`
`Phase Transitions During Lyophilization and Storage
`
`During initial cooling of protein formulation solutions, water is normally the fi rst
`component to crystallize. At this stage, a biphasic system is formed, consisting of
`ice and residual freeze-concentrated solution (FCS), which contains protein drug,
`excipients, and remaining water. The composition of the FCS after initial (also
`known as primary) water crystallization depends on the ratio of solutes and the
`temperature, but is independent of total solid content (11). As cooling (and water
`crystallization) proceeds further, the FCS may either remain in the amorphous
`state or partially crystallize, depending on the composition of the system and the
`cooling rate (11) as described below:
`
`1. The FCS may form a kinetically stable (but thermodynamically unsta-
`ble) amorphous phase. A typical example of such behavior is sucrose-
`rich formulations. In this case, solutes do not usually crystallize during
`freezing, drying, and storage, provided that the freeze-dried cake is
`protected from water uptake and the storage temperature is well below
`the glass transition temperature.
`2. The FCS may form a “doubly unstable” (i.e., both thermodynamically
`and kinetically unstable) state. Mannitol- and glycine-based formula-
`tions are typical examples of such behavior. In these systems, second-
`ary excipient + ice crystallizationa would occur either during cooling (if
`the cooling rate is slower than the critical cooling rate) or subsequent
`heating/annealing of the frozen solution (if the cooling rate was higher
`than the critical cooling rate). The critical cooling rate depends on the
`composition of the solution (e.g., glycine/sucrose ratio) and increases
`with an increase in the fraction of a crystallizable component (12). It
`should be noted that crystallization of an excipient is often incomplete,
`i.e., the maximal FCS contains usually all the components (water and
`all the solutes including protein and the excipients), although the rela-
`tive fraction of the partially crystallized excipient remaining in the FCS
`is signifi cantly reduced.
`
`In addition to the common cases described above, it was proposed that a liquid–
`liquid (amorphous–amorphous) phase separation might take place, resulting in
`two amorphous phases of different chemical composition (3,9,13). For example,
`
`a Note also that the secondary solute+water crystallization is referred in the pharmaceutical literature
`as “eutectic” crystallization, although this is not a strictly correct term to apply to a multicomponent
`system.
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`200
`
`Shalaev et al.
`
` protein–excipient amorphous–amorphous phase separation would result in pro-
`tein-rich and excipient-rich amorphous phases. Such phase separation is expected
`to compromise the stabilizing activity of excipients. It should be stressed, however,
`that there is a lack of experimental reports on such demixing behavior between
`protein and amorphous excipient during lyophilization, and it is not clear if this is
`a common behavior for protein formulations.
`Although the phase state of excipients is usually “fi xed” during freezing
`and annealing, further phase transformations may take place during primary and
`secondary drying as well as during shelf storage, depending on the properties and
`the concentration of excipients as well as the storage temperature. For example,
`if an excipient crystallizes as a crystallohydrate (i.e., a crystal with water in the
`crystal lattice) during freezing, the water of hydration might be removed dur-
`ing either primary or secondary drying. Such removal of water of hydration can
`result in either amorphous [e.g., sodium phosphate (14)], or crystalline anhydrous
`(e.g., mannitol) excipient. In addition, an amorphous excipient [e.g., inositol (15)]
`may crystallize during the shelf-life, especially if the water content in the lyocake
`increased because of water transfer either from the stopper or (if the stopper was
`not properly sealed) from the environment.
`
`Signifi cance of Excipient Crystallization
`
`Phase transitions of excipients during manufacturing and storage have a major
`impact on both stability and performance of protein dosage forms. In particular,
`crystallization of either a buffer or lyo- and a cryoprotector is usually undesir-
`able because of the negative impact on protein stability. Indeed, crystallization of
`buffer components is often accompanied by signifi cant changes in the pH of the
`FCS (16), which often causes destabilization of a protein. Also, crystallization
`of a lyoprotector can compromise its protective function. For example, it was
`shown that inositol stabilized a protein when it existed as an amorphous form,
`whereas loss of protein activity was observed when inositol crystallized during
`storage (15). Another example is crystallization of a lyoprotector, raffi nose, dur-
`ing freezing, causing destabilization of lactate dehydrogenase (LDH) (17). The
`negative impact of a lyoprotector (e.g., sugar) crystallization on protein stability
`can be attributed to two different factors: (i) Crystallization results in a physi-
`cal separation of sugar molecules from protein molecules, i.e., an increase in
`intermolecular sugar/protein distance, from several angstroms [which is a typical
`hydrogen-bond length (18)] in molecular mixtures to micrometers in physical
`mixtures of crystalline sugar and amorphous protein; such separation would be
`expected to eliminate any protection imparted by sugars irrespective of the exact
`mechanism (e.g., water substitution vs. the glass transition hypothesis, or ther-
`modynamics vs. kinetics mechanism). (ii) Crystallization of a sugar in an anhy-
`drous form would result in a redistribution of water and in a signifi cant increase
`in the local water content of the remaining amorphous protein-containing phase,
`which could be detrimental to long-term stability. It should be noted, however,
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`Rational Choice of Excipients for Use in Lyophilized Formulations
`
`201
`
`that a sugar crystallohydrate (e.g., a pentahydrate as with raffi nose) might serve
`as a water “scavenger” during crystallization, thus preventing an increase in local
`water content (19,20).
`From the manufacturing process point of view, however, crystallization
`of an excipient may be benefi cial because it allows primary drying at a higher
`temperature without visible collapse, which results in a shorter and more robust
`freeze-drying cycle. In addition, partially crystalline formulations have higher
`drying rates (i.e., shorter cycle) than amorphous formulations of a similar compo-
`sition, possibly because a higher fraction of water is isolated as ice, with ice easier
`to remove than nonfrozen water associated with the amorphous phase (11).
`A compromise between a desire to improve freeze-drying cycle effi ciency
`and robustness (which is achieved by using a crystalline bulking agent) and
`sustaining protein protection (which needs an amorphous lyoprotector) can be
`achieved by using partially crystalline–partially amorphous formulations (21). It
`has been proposed that the crystalline portion provides a physical support even
`at relatively high product temperatures (i.e., higher than the collapse temperature
`of the amorphous phase) whereas the amorphous portion provides lyoprotection
`for protein, allowing aggressive primary drying conditions (21). Feasibility of
`such crystalline–amorphous formulations was demonstrated using glycine–sugar
`formulations with a lyophilization-sensitive enzyme, LDH (22). In this system,
`freeze-drying at a product temperature more than 10°C above the Tg′ resulted
`in a freeze-dried cake without any evidence of macroscopic collapse and with a
`retained enzymatic activity, when the crystalline/amorphous ratio was higher than
`1.2/1 (raffi nose) or 1.6/1 (trehalose) (22). One should be aware, however, that
`timing of crystallization of a crystalline bulking agent, i.e., whether the crystalli-
`zation takes place during cooling or annealing, may infl uence protein stability. An
`example of the signifi cance of crystallization conditions was given in Ref. (11),
`where the stability of a freeze-dried conjugate of immunoglobulin G and horse-
`radish peroxidase in a partially crystalline glycine/sucrose matrix was reported.
`In this case, the activity recovered was signifi cantly higher in the material that
`crystallized during annealing as compared with material in which crystallization
`occurred during cooling. Therefore, although crystalline–amorphous formula-
`tions may be benefi cial, the phase behavior and protein stability need to be inves-
`tigated in each particular case in order to ensure a stable and robust freeze-dried
`product.
`Amorphous–amorphous (liquid–liquid) phase separation of excipients also
`may cause protein destabilization, probably because of creation of an interface
`(1). In addition, it is also possible that a similar amorphous– amorphous demixing
`may occur between protein and lyoprotectant, with expected loss of protection
`(4,9), although there is a lack of experimental data on such protein–excipient
`amorphous–amorphous phase separation. Note that excipient– excipient demix-
`ing in FCS can be studied by differential scanning calorimetry DSC (23), whereas
`no reliable methods to detect such transitions exist for protein–lyoprotectant
` amorphous–amorphous demixing.
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`202
`
`Shalaev et al.
`
`Phase State of Excipients: Analytical Aspects
`
`Excipients can undergo phase transitions during different stages of freeze-drying
`and over the shelf-life of the freeze-dried product. To develop a robust and stable
`formulation, it is essential to understand and monitor such changes. The most
`common methods to detect crystalline–amorphous changes are DSC and X-ray
`diffraction (XRD), whereas other methods such as polarized light microscopy
`(PLM) and different spectroscopic techniques can also be used, depending on the
`formulation properties and stage in the formulation “life” when such transitions
`may occur.
`Excipient crystallization may usually be expected to occur during the freez-
`ing and annealing stages of freeze-drying. Such events are commonly and con-
`veniently studied by DSC. On a DSC curve, crystallization can be detected as a
`second exotherm during cooling (with a fi rst exotherm being water crystalliza-
`tion), an exotherm during heating, and/or an additional endotherm during heating
`preceding the main ice melting peak. If either of these events is observed on DSC
`cooling–heating curves, one may conclude that a component(s) of the formulation
`would likely crystallize during freeze-drying. The reverse statement, however,
`is not always correct, i.e., lack of a crystallization event in a DSC experiment does
`not necessarily mean that crystallization would not occur in vials during freeze-
`drying. Indeed, relatively high scanning rates and small sample volume in a DSC
`study would provide less favorable crystallization conditions as compared with a
`larger sample volume and slower temperature ramping during a real freeze-drying
`run. Low-temperature XRD is another main method used to study crystalliza-
`tion in aqueous solutions (12,24), and is especially well suited both to distinguish
`between crystalline and amorphous structures and to identify the nature of any
`crystalline phase(s) present.
`X-ray powder diffraction (XRPD) is probably the most common and conve-
`nient method to detect crystalline structures in a freeze-dried cake. PLM can also
`be used to confi rm the amorphous nature of a freeze-dried cake. If birefringence is
`observed, it usually means that the cake is at least partially crystalline. However,
`PLM does not usually allow for the identifi cation of the specifi c crystalline phase.
`Note that a sample is often exposed to an ambient atmosphere during both XRPD
`and PLM experiments, which may result in water uptake followed by crystalliza-
`tion. Therefore, precautions should be taken to minimize sample exposure to ambi-
`ent relative humidity (RH) during measurements, to prevent erroneous conclusions.
`As indicated previously, DSC is another common tool to distinguish between
`crystalline and amorphous formulations. Spectroscopic methods such as Fourier
`transform infrared (FTIR) and Raman, and solid state nuclear magnetic resonance
`(NMR) may also be used for structure characterization of lyophile cakes.
`Confi rmation of the amorphous nature of a formulation is essential during the
`formulation development process, e.g., when a formulator needs to choose a buffer
`and/or lyoprotector. For both buffer and lyoprotector, retention of the excipient in
`an amorphous state is desirable, and can serve as a key criterion for the selection
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`Rational Choice of Excipients for Use in Lyophilized Formulations
`
`203
`
`of an excipient. Usually, a combination of evaluation of the pre-lyo solution using
`DSC and evaluation of the fi nished cake using XRPD is suffi cient to conclude if
`any crystallization would occur during freeze-drying. For example, if no crystal-
`lization is observed by DSC and a freeze-dried cake is amorphous by X-ray, one
`could conclude that no crystallization of either buffer or lyoprotector occurred dur-
`ing lyophilization. However, although the solution–DSC/freeze-dried-cake–XRPD
`combination is usually suffi cient to make a reliable conclusion about excipient(s)
`crystallization, it is not always the case. In particular, an erroneous conclusion
`might be made in cases when the following conditions are met: (i) an excipient
`crystallizes as a hydrate; (ii) water of hydration is removed during drying (e.g.,
`secondary drying); (iii) loss of water of hydration causes crystal-to-amorphous
`conversion. Although it might appear that such a combination of events is unlikely,
`it was shown by in situ freeze-drying XRD that this scenario can take place in real
`systems, such as phosphate buffer (14) and the lyoprotectant raffi nose (17). As
`a result of such fi ndings, use of the in situ XRPD method is attracting increased
`attention in the fi eld. In addition, it is important to note a recent improvement
`in XRPD through the use of a high-intensity synchrotron radiation source, which
`provides an excellent signal-to-noise ratio and superior sensitivity as compared to
`the traditional XRPD method. Use of synchrotron XRPD (sXRPD) is especially
`important when one needs to detect crystallization of a low-concentration excipi-
`ent, e.g., buffer, where sensitivity is a major issue. Application of sXRPD in the
`analysis of both freeze-dried cakes and phase transitions in frozen solutions and
`during freeze-drying can be found elsewhere (25,26).
`
`ROLE AND PROPERTIES OF EXCIPIENTS
`
`Buffers
`
`Control of pH is often needed to ensure optimal solubility and stability of a prod-
`uct during manufacturing, storage, and upon reconstitution. In most cases, an
`appropriate amount of buffer is needed to provide adequate buffering capacity.
`Buffer type and concentration, as well as solution pH before lyophilization, are
`important formulation variables. Buffering capacity and the possibility of buffer
`catalysis are the major buffer properties to be considered in the development of
`liquid pharmaceutical formulations (27). There are additional requirements for
`buffers for freeze-drying, i.e., they should be nonvolatile, have a high collapse
`temperature (Tc or Tg′) in the FCS, remain amorphous during freeze-drying, and
`have a high glass transition temperature in the solid state (28). Several buffers
`that are common for parenteral formulations have unfavorable freeze-drying
`properties. For example, acetate—a common buffer—is not a preferred buffer for
`lyophilization because it is volatile and can be partially lost during freeze-drying,
`resulting in a signifi cant pH change. Hydrochloric acid is another example of a
`pH modifi er that is volatile and should be used with caution. In addition, sev-
`eral common buffers have a high tendency to crystallize during freezing. Buffer
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`204
`
`Shalaev et al.
`
`crystallization is usually undesirable because it can lead to substantial pH shifts
`during freezing and therefore could destabilize the protein. In particular, sodium
`phosphate buffer demonstrated signifi cant pH changes (several pH units) during
`freezing, as a result of freeze concentration and crystallization of the buffer com-
`ponents (16). Similarly, tartrate and succinate buffers crystallize readily whereas
`citrate, glycolate, and malate are more resistant to crystallization (29). It should
`be noted that both crystallization and collapse behavior depend on solution pH.
`For example, collapse temperatures of several common buffers are presented in
`Figure 1 as a function of solution pH. The fi gure illustrates two interesting fea-
`tures of the collapse behavior of buffers, i.e., signifi cant changes in the collapse
`temperature with solution pH, and an infl uence of a counter ion (e.g., sodium
`citrate vs. potassium citrate).
`Overall, buffers with a higher collapse temperature (Tc or Tg′) and a lower
`crystallization potential are preferred for lyophilized formulations. However, it
`should be stressed that buffers with a relatively low collapse temperature and
`relatively high crystallization potential (e.g., phosphate buffer) can still be used
`in lyophilized formulations. Both collapse temperature and crystallization ability
`can be modifi ed using other excipients. For example, if a formulation contains a
`
`Figure 1 Collapse temperature (Tg′) as a function of pH. n: sodium citrate; ®: potassium
`citrate; ∆: sodium tartrate; ´: L-histidine. Error bars represent standard deviation. Lines are
`given as a visual help. Source: From Refs. 29, 30.
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`Rational Choice of Excipients for Use in Lyophilized Formulations
`
`205
`
`signifi cant amount of a solute with a high Tc (e.g., a protein), the collapse tem-
`perature of the formulation would increase and buffer crystallization may be sup-
`pressed. However, the amount of an amorphous component required to suppress
`crystallization and/or to provide an acceptable Tc would be lower in the case of
`a buffer with a higher Tc or with a lower crystallization potential. Citrate buf-
`fer appears to be a good choice for lyophilized formulations that are prepared at
`acidic or near neutral pH. Frozen solutions of citrate buffer have a low crystal-
`lization potential, relatively high collapse temperature, and minimal pH changes
`during freezing. In addition, citrate has a reasonably high Tg′ especially when
`prepared from solutions at higher pH values. Glycine may be a reasonable choice
`for the alkaline pH region based on a relatively low crystallization potential of
`sodium glycinate. Other details on freeze-drying properties of different buffers
`can be found in Ref. (28).
`
`Bulking Agents/Lyoprotectors
`
`In many cases, the dose of a drug is quite small, and a bulking agent (or fi ller) is
`needed to provide a matrix to carry the active ingredient. Common bulking agents
`include mannitol, lactose, sucrose, dextran, trehalose, and glycine. In protein for-
`mulations, bulking agents often play a dual role as both bulking agent and lyopro-
`tector. These bulking agents range both in their ability to crystallize (mannitol and
`glycine) or remain amorphous (e.g., sucrose) and in their impact on the formula-
`tion collapse or eutectic temperature. An appropriate choice of a bulking agent
`results in optimal product quality (e.g., physical and chemical stability, reconstitu-
`tion time, moisture levels) and facilitates freeze-drying and scale-up to production
`size. The level of bulking agent utilized will vary depending on the rationale for
`use, e.g., as a matrix-forming agent, as the collapse temperature modifi er, or stabi-
`lizer. As a “rule of thumb,” use of a bulking agent can be considered if the active
`concentration in the fi ll solution is less than 2 wt%.
`Mannitol is the most common bulking agent used in protein freeze-dried
`formulations. Usually, mannitol crystallizes during cooling or annealing of fro-
`zen solutions, which, combined with a high mannitol–ice eutectic temperate
`of −1.5°C (31), allows one to freeze-dry such formulations at a relatively high
`primary drying temperature, without a macroscopic collapse. Therefore, manni-
`tol-based formulations are known to be easy to lyophilize, with a shorter and
`robust lyophilization cycle. There are two potential complications associated with
` mannitol-based formulations. Mannitol forms a crystallohydrate during freeze-
`drying (32,33) that hinders removal of water of hydration and requires elevated
`secondary drying temperature. Vial breakage is another potential problem a for-
`mulator might encounter while working with mannitol-rich formulations (34,35).
`It has been shown that vial breakage is likely associated with volume expansion
`that occurs during warming of a frozen solution at approximately −25°C to −20°C
`(36) probably because of secondary mannitol + water crystallization. Vial break-
`age is affected by mannitol concentration, cooling rate, and fi lling volume (34),
`
`

`

`McNally
`
` PTR
`
` 09/24/07
`
` Chapter 09
`
`206
`
`Shalaev et al.
`
`as well as presence of amorphous solutes and vial confi guration (35). Glycine is
`another common crystalline bulking agent. Because of ionizable groups of gly-
`cine, it may also serve as a buffer. At neutral pH, glycine crystallizes as anhydrous
`beta polymorph, and its eutectic with ice has a relatively high eutectic temperature
`of −3.6°C (37). Both solution pH and counter ions have a signifi cant impact on
`glycine crystallization behavior (38), by infl uencing both ionization of glycine
`in solution and a precipitating solid form. Usually, the presence of other solutes
`(such as a lyoprotector or the protein itself) hinders crystallization of both glycine
`and mannitol. For example, sucrose inhibited the crystallization of mannitol at a
`sucrose/mannitol ratio of 2:1 (39). Inhibition of glycine crystallization by sugars
`(sucrose, trehalose, and raffi nose) was reported in Refs. (12,40,41). For example,
`a critical sucrose/(glycine + sucrose) ratio above which glycine does not crystal-
`lize during either cooling or annealing was reported to be 0.8 (12).
`Sucrose is probably the most popular lyoprotector used in lyophile pro-
`tein formulations. Because of its low collapse temperature, however, lyophilizing
`sucrose-rich formulations can be a challenge. In addition, the presence of other
`components with low collapse temperatures (e.g., buffers) may lower the Tc even
`further. Possible ways to overcome this challenge are reducing the concentration
`of components with low Tc, addition of an excipient with a high collapse tempera-
`ture (e.g., dextran), or addition of a crystalline bulking agent. A critical factor for
`the selection of a lyoprotector is its impact on physical and chemical stability of
`proteins, both during freeze-drying and the product shelf-life. Sucrose is known to
`substantially increase the stability of proteins both during freeze-drying and dur-
`ing subsequent storage. Lactose is another popular bulking agent although there
`is a potential for chemical interaction with amino groups of a protein forming a
`Schiff base, known as the Maillard reaction, or nonenzymatic browning. Recently,
`another disaccharide lyoprotector was introduced with claims of superior stabili-
`zation properties, i.e., trehalose (42). The most obvious advantage of trehalose is
`its higher glass transition temperature as compared to sucrose (120°C vs. 74°C).
`However, side-by-side comparison of stabilization of proteins by sucrose and tre-
`halose revealed that sucrose might provide a comparative or, in some cases, better
`protection than trehalose. For example, sucrose appeared to be more effective in
`stabilizing the native structure of lysozyme during spray

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