throbber
Pharmaceutical Biotechnology - Volume 13
`
`
`
`ational DeSIg
`of Stable Protel
`Formulations
`Theoryand Practice
`
`
`
`
`
`Edited by
`John F. Carpenter
`'
`' and
`Mark C. Manning
`
`MAIA Exhibit 1011
`MAIA V. BRACCO
`
`,- IPR PETITION
`
`
`
`
`MAIA Exhibit 1011
`MAIA V. BRACCO
`IPR PETITION
`
`

`

`Rational Design of Stable
`Protein Formulations
`Theory and Practice
`
`
`
`
`

`

`Rational Design of Stable
`Protein Formulations
`Theory and Practice
`
`
`
`
`

`

`Pharmaceutical Biotechnology
`
`Series Editor: Ronald T. Borchardt
`The University of Kansas
`Lawrence, Kansas
`
`Recent volumes in this series:·
`
`Volume 7
`
`Volume 8
`
`PHYSICAL METHODS TO CHARACTERIZE
`PHARMACEUTICAL PROTEINS
`Edited by James N. Herron, Win Jiskoot,
`and Daan J. A. Crommelin
`
`MODELS FOR ASSESSING DRUG ABSORPTION
`AND METABOLISM
`Edited by Ronald T. Borchardt, Philip L. Smith,
`and Glynn Wilson
`
`Volume 9
`
`FORMULATION, CHARACTERIZATION, AND
`STABILITY OF PROTEIN DRUGS: Case Histories
`Edited by Rodney Pearlman and Y. John Wang
`
`Volume 10 PROTEIN DELIVERY: Physical Systems
`Edited by Lynda M. Sanders and R. Wayne Hendren
`
`Volume 11
`
`INTEGRATION OF PHARMACEUTICAL DISCOVERY AND
`DEVELOPMENT: Case Histories
`Edited by Ronald T. Borchardt, Roger M. Freidinger,
`Tomi K. Sawyer, and Philip L. Smith
`
`Volume 12 MEMBRANE TRANSPORTERS AS DRUG TARGETS
`Edited by Gordon L. Amidon and Wolfgang Sadee
`
`Volume 13 RATIONAL DESIGN OF STABLE PROTEIN
`FORMULATIONS: Theory and Practice
`Edited by John F. Carpenter and Mark C. Manning
`
`Volume 14 DEVELOPMENT AND MANUFACTURE OF PROTEIN
`PHARMACEUTICALS
`Edited by Steven L. Nail and Michael J. Akers
`
`A Chronological Listing of Volumes in this series appears at the back of this volume
`
`A Continuation Order Plan is available for this series. A continuation order will bring delivery of each
`new volume immediately upon publication. Volumes are billed only upon actual shipment. For further
`information please contact the publisher.
`
`
`
`
`

`

`Rational Design of Stable
`Protein Formulations
`Theory and Practice
`
`Edited by
`John F. Carpenter
`and
`Mark C. Manning
`University of Colorado Health Sciences Center
`'Denver, Colorado
`
`uwer Academic I Plenum Publishers
`w York, Boston, Dordrecht, London, Moscow
`
`~.:
`
`'1;'t
`
`
`
`
`

`

`Library of Congress Cataloging-in-Publication Data
`
`Rational design of stable protein formulations: theory and
`practice/edited by John F. Carpenter, Mark C. Manning.
`p.
`cm. -
`(Pharmaceutical biotechnology; v. 13)
`Includes bibliographical references and index.
`ISBN 0-306-46741-0
`1. Protein drugs-Stability. 2. Protein engineering. 3.
`I. Carpenter, John F.
`Drugs-Design.
`II. Manning, Mark C.
`Series.
`RS431.P75 R38 2002
`615' .19-dc21
`
`ill.
`
`2001057997
`
`ISBN: 0-306-46741-0
`
`© 2002 Kluwer Academic/.Plenum Publishers, New York
`233 Spring Street, New York, N.Y. 10013
`
`http://www.wkap.nl/
`
`109876543 21
`
`A C.I.P. record for this book is available from the Library of Congress
`
`All rights reserved
`
`No part of this book may be reproduced, stored in a retrieval system, or transmitted in any form
`or by any means, electronic,_ mechanical, photocopying, microfilming, recording, or otherwise,
`without written permission from the Publisher
`
`Printed in the United States of America
`
`
`
`
`

`

`Contents
`
`Chapter 1
`
`Practical Approaches to Protein Formulation Development
`Byeong S. Chang and Susan Hershenson
`
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Preparation for Formulation Development ..................... .
`Resource Requirements for Formulation Development ... : ...... .
`Useful Information for Designing Formulations ............... .
`Preformulation Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Characterization of Protein Pharmaceuticals .................. .
`Accelerated Stability Studies ............................. .
`Development of Analytical Methods ....................... .
`Evaluation of the Significance of Problems .................. .
`Formulation Development ................................. .
`Formulation Options for Protein Pharmaceuticals ............... ·
`Typical Protein Stability Problems: Causes and Solutions ........ .
`Optimization of Formulation Variables ...................... .
`Necessary Studies for Formulation Development .............. .
`Strategies to Overcome Difficult Formulation Problems ......... .
`Fonnulation in Commercial Product Development ............... .
`Critical Formulation Decisions During Pharmaceutical
`·Development ........................................ .
`Formulation for Early Preclinical and Clinical Studies .......... .
`Commercial Formulation ................................ .
`Regulatory Issues in Formulation Development ............... .
`
`1
`3
`3
`4
`4
`5
`5
`6
`7
`10
`10
`13
`13
`15
`17
`18
`
`18
`19
`19
`20
`
`xiii
`
`I
`
`1,
`
`I
`:1
`
`
`
`
`

`

`xiv
`
`Contents
`
`. . . . . . . . . . . . . . . . . . . . . .
`Appendix: List of Regulatory Documents
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`22
`23
`
`Chapter 2
`
`Recombinant Production of Native Proteins from Escherichia coli
`Tsutomu Arakawa, Tiansheng Li, and Linda 0. Narhi
`
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Distribution of Expressed Proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Cell Washing and Lysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Purification of Soluble, Folded Proteins . . . . . . . . . . . . . . . . . . . . . . . .
`Purification and Refolding of Soluble, Misfolded Proteins
`. . . . . . . . . .
`Purification and Refolding of Proteins from Inclusion Bodies . . . . . . . .
`Washing and Solubilization of Inclusion Bodies . . . . . . . . . . . . . . . .
`Purification of Expressed Proteins from Inclusion Bodies . . . . . . . . .
`Refolding Mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Disulfide Bond Formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Removal of Denaturant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Effects of Tag Sequences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Effects of Excipients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Response Surface Methodology . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`High Pressure Disaggregation and Refolding
`. . . . . . . . . . . . . . . . . .
`Methods to Analyze Folded Structures . . . . . . . . . . . . . . . . . . . . . . . . . .
`Bioactivity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Binding to Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Dilsulfide Bond Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Spectroscopy
`Conformational Stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Limited Proteolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`27
`28
`32
`34
`35
`36
`36
`36
`38
`41
`41
`44
`44
`47
`48
`48
`49
`49
`50
`50
`51
`51
`51
`
`Chapter 3
`
`Physical Stabilization of Proteins in Aqueous Solution
`Brent S. Kendrick, Tiansheng Li, and Byeong S. Chang
`
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Overview of Physical Stability
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`61
`62
`
`
`
`
`

`

`Contents
`
`Thermodynamic Control of Protein Stability ................. .
`Kinetic Control of Protein Stability ........................ .
`Interactions of Excipients with Proteins ....................... .
`Preferentially Excluded Cosolvents ........................ .
`Buffers/Salts ......................................... .
`Specific Binding of Ligands ............................. .
`Protein Self-Stabilization ................................ .
`Physical Factors Affecting Protein Stability .................... .
`Temperature .......................................... .
`Freeze-Thawing ...................................... .
`Agitation and Exposure to Denaturing Interfaces .............. .
`Pressure ..... · ....................................... .
`Conclusions ........................................... .
`Appendix: Derivation of the Wyman Linkage Function and
`Application to the Timasheff Preferential Exclusion
`Mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References
`
`xv
`
`62
`63
`65
`66
`67
`68
`69
`70
`70
`71
`71
`72
`73
`
`73
`78
`
`Chapter 4
`
`Effects of Conformation on the Chemical Stability of Pharmaceutically
`Relevant Polypeptides
`Jeffrey D. Meyer, Bert Ho, and Mark C. Manning
`
`Introduction ... ·. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Relationship Between Structure and Deamidation Rates . . . . . . . . . . . .
`Primary Structure Effects
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Secondary Structure Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Tertiary Structure Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Summary of Structure Effects on Deamidation . . . . . . . . . . . . . . . . .
`Role of Structure in Protein Oxidation . . . . . . . . . . . . . . . . . . . . . . . . .
`Types of Oxidation Processes
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Effects of Oxidation of Surface and Buried Methionines on
`Protein Structure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Limiting Solvent Accessibility of Residues . . . . . . . . . . . . . . . . . . . .
`Conformational Control of Oxidation in Aqueous Solution . . . . . . . .
`Structural Control of Oxidation in Lyophilized Products . . . . . . . . . .
`Summary of Structural Control of Oxidation
`. . . . . . . . . . . . . . . . . .
`Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References . . . . . . . . , . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`85
`86
`87
`89
`91
`92
`92
`93
`
`95
`96
`97 .
`99
`100
`101
`101
`
`
`
`
`

`

`xvi
`
`Chapter 5
`
`Contents
`
`Rational Design of Stable Lyophilized Protein Formulations:
`Theory and Practice
`John F. Carpenter, Beyong S. Chang, William Garzon-Rodriguez, and
`Theodore W. Randolph
`
`109
`111
`112
`
`113
`114
`114
`
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Minimal Criteria for a Successful Lyophilized Formulation
`. . . . . . . . .
`Inhibition of Lyophilization-Induced Protein Unfolding
`. . . . . . . . . .
`Storage at Temperatures Below Formulation Glass Transition
`Temperature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`The Water Content is Relatively Low . . . . . . . . . . . . . . . . . . . . . . . .
`A Strong, Elegant Cake Structure is Obtained . . . . . . . . . . . . . . . . . .
`Steps Taken to Minimize Specific Routes of Protein Chemical
`Degradation ............................... : . . . . . . . .
`Rational Design of Stable Lyophiilized Formulations . . . . . . . . . . . . . .
`Choice of Buffer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Specific Ligands/pH that Optimizes Thermodynamic Stability of
`Protein ................ , . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Trehalose or Sucrose to Inhibit Protein Unfolding and Provide
`Glassy Matrix ...... _. ;. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
`Bulldng Agent (e.g., Mannitol, Glycine or Hydroxyethyl Starch) . . . .
`126
`Nonionic Surfactant to Inhibit Aggregation. . . . . . . . . . . . . . . . . . . .
`127
`Acknowledgments
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`127
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`127
`
`116
`117
`118
`
`119
`
`Chapter 6
`
`Spray-Drying of Proteins
`Geoffrey Lee
`
`. . . . . . . . . . . . . . . . . . . . . . .
`Introduction: Why Spray-Dry a Protein?
`Developments in the Last 10 Years
`. . . . . . . . . . . . . . . . . . . . . . . . . . .
`The Practice of Spray-Drying Proteins . . . . . . . . . . . . . . . . . . . . . . . . .
`Type of Equipment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Spray-Drying Conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Influence of Formulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Pure Proteins
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Formulated Systems
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Use of Added Surface Active Substances . . . . . . . . . . . . . . . . . . . . .
`
`135
`136
`139
`139
`140
`147
`14 7
`149 ·
`151
`
`
`
`
`

`

`Contents
`
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Concluding Remarks
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`xvii
`
`156
`156
`
`Chapter 7
`
`Surfactant-Protein Interactions
`Theodore W Randolph and LaToya S. Jones
`
`Introduction .................................... ; . . . . . . .
`Proteins and Surfactants at Surfaces
`. . . . . . . . . . . . . . . . . . . . . . . . . .
`Protein-Surfactant Interactions irt Solution . . . . . . . . . . . . . . . . . . . . . .
`Surfactant Effects on Protein Assembly State . . . . . . . . . . . . . . . . . . . .
`Surfactant Effects-on Proteins During Freezing, Freeze-Drying
`and Reconstitution
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Enzymatic Degradation of Non-Ionic Surfactants . . . . . . . . . . . . . . . . .
`Recommendations for Protein Formulation
`. . . . . . . . . . . . . . . . . . . . .
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`159
`161
`166
`167
`
`169
`170
`170
`171
`
`Chapter 8
`
`·High Throughput Formulation: Strategies for Rapid Development of
`Stable Protein Products
`Rajiv Nayar and Mark C. Manning
`
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Overall Structure of the RTF Approach ....................... .
`Role of an Established Decision Tree for Formulation Design ....... .
`Constraints on a Pharmaceutically Acceptable Protein
`Formulation ....................................... .
`Proper Choice of Dosage Form ........................... .
`Preformulation Studies ................................. .
`Proper Choice of Excipients
`. . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Estimates of Resources Needed for Formulation Development .... .
`Use of Software and Databases to Assist in the RTF Process ....... .
`Essential Analytical Methods .............................. .
`Stability Protocols .................................... · · ·
`Unified Strategy for RTF ................................. .
`References . . . . . . . . . . . . . . . . . . . . . · · · · · · · · · · · · · · · · · · · · · · · ·
`
`hidex ................................................ .
`
`177
`179
`181
`
`182
`183
`185
`186
`188
`189
`191
`193
`194
`195
`
`199
`
`\\ ,,
`
`
`
`
`

`

`5
`
`Rational Design of Stable
`Lyophilized Protein Formulations:
`Theory and Practice
`
`John F. Carpenter1,2, Beyong S. Chanl,
`2
`William Garzon-Rodriguez1
`, and
`•
`Theodore W. Randolph1
`,4
`
`INTRODUCTION
`
`For ease of preparation and cost containment by the manufacturer, and ease of
`handling by the end user, an aqueous therapeutic protein formulation usually is
`preferred. However, with many proteins it is not possible-especially consider(cid:173)
`ing the time constraints for product development-to develop sufficiently stable
`aqueous formulations. Unacceptable de11aturation and aggregation can be induced
`readily by the numerous stresses to which a protein in aqueous solution is sensi(cid:173)
`tive; e.g., heating, agitation, freezing, pH changes, and exposure to interfaces or
`denaturants (Arakawa et al., 1993; Cleland et al., 1993; Brange, 2000; Bummer
`
`John F. Carpenter, William Garzon-Rodriguez, and Theodore W. Randdph
`• Center for Phar(cid:173)
`John F. Carpenter, William Garzon-Rodriguez, and Theodore W.
`maceutical Biotechnology.
`• Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado
`Randolph
`• Amgen, Inc., Thousand
`Beyong S. Chang
`Health Sciences Center, Denver, CO 80262.
`Theodore W. Ranqolph
`• Department of Chemical Engineering, Univer(cid:173)
`Oaks, CA 91320.
`sity of Colorado, Boulder, CO 80309.
`Rational Design of Stable Protein Formulations, edited by Carpenter and Manning. Kluwer Academic I Plenum
`Publishers, New York, 2002.
`
`109
`
`
`
`
`

`

`110
`
`John F. Carpenter et al.
`
`and Koppenol, 2000). Furthermore, even under conditions that thermodynami(cid:173)
`cally greatly favor the native state of proteins, aggregation can arise during
`months of storage in aqueous solution (e.g., Gu et al., 1991; Arakawa et al., 1993:
`Chen et al., 1994; Volkin and Middaugh, 1996; Chang et al., 1996a). In addition.
`several chemical degradation pathways (e.g., hydrolysis and deamidation) are
`mediated by water. In aqueous formulations, the rates of these and other (e.g ..
`oxidation) chemical degradation reactions can be unacceptably rapid on the time
`scale of storage (e.g., 18-24 months) for pharmaceutical products (Manning e1
`al., 1989; Cleland et al., 1993; Goolcharran et al., 2000; Bummer and Koppenol
`2000).
`In contrast, a properly lyophilized formulation can maintain adequate phys·
`ical and chemical stability of the protein during shipping and long-term storage
`even at ambient temperatures. As will be outlined in this chapter, developin§
`stable lyophilized protein formulations should be a rational, straightforwarc
`process, which for most proteins should be rapid. With liquid formulation devel·
`opment, it may only be possible to obtain adequate protein stability after length)
`studies. Furthermore, sometimes there are conflicting conditions (e.g., pH:
`needed to slow sufficiently multiple degradation pathways in aqueous solution
`Considering these issues plus the fact that formulation scientists now have to dea
`with numerous proteins and/or variants of a given protein, lyophilization shoulc
`be considered as a primary mode for product development. Only if a paralle
`effort to develop an aqueous formulation is successful, will a final lyophilizec
`product not be needed.
`Rapid formulation development has important financial ramifications. P
`drug product has a finite patent life, during which time the company has an exclu·
`sive market. Considering that even a moderately successful drug product ha~
`annual sales of hundreds of millions of dollars, potentially millions of dollars ir
`sales are lost for each day of delay in bringing a product to market. Unfortunately
`there are often delays because the formulations designed during early stage devel
`opment and clinical trials (e.g., frozen) were not adequate for the final product
`With a rational approach to formulation development, pharmaceutical scientist:
`and process engineers can minimize the risk of this problem and the time neede<
`to obtain a successful, final formulation. The success of such efforts depends or
`frank, open communication between the groups involved. For example, it is crit
`ical that the formulation scientists learn from the process engineers the issues fo:
`large-scale lyophilization tuns, which are usually conducted in units that do no
`have the capacity to match processing parameters obtained· in a small-seal<
`research lyophilizers.
`Despite the best efforts of the scientists and engineers, all too often delay:
`in formulation development arise because sufficient resources are not invested it
`product development. For example, sometimes, purchase of essential equipmen
`
`---------------- .
`
`
`
`
`

`

`II:
`I ,,
`
`Rational Design of Stable Lyophilized Protein Formulations
`
`111
`
`(e.g., a differential scanning calorimeter), which costs a minute fraction of a day's
`sale of product, is not allowed. To avoid unnecessary delays in product launch,
`which can have disastrous consequences for the company and for patients, it is
`essential that companies appreciate that product development is ultimately a key
`limiting factor in getting a therapeutic to market. Hence, development efforts need
`to be as well funded as the usually much more visible drug discovery research
`programs. If a product is not stable, it will not be marketed, no matter how dra(cid:173)
`matic an impact it can have on human health and the financial status of the
`company.
`
`MINIMAL CRITERIA FOR A SUCCESSFUL
`LYOPHILIZED FORMULATION
`
`Research over the past several years has demonstrated that five criteria
`define the minimal conditions necessary for a successful lyophilized protein for(cid:173)
`mulation (Table 1).
`The first four criteria can be met with use of the appropriate excipients and
`lyophilization cycle design. For information on the proper design of lyophiliza(cid:173)
`tion cycles, the reader is directed to the numerous previous reviews in this
`area (Franks, 1990; Pikal, 1990; Nail and Gatlin, 1993; Gatlin and Nail, 1994;
`Carpenter and Chang, 1996; Rey and May, 1999; Cappola, 2000). For the current
`chapter we will only consider cycle design in terms of the interplay between for(cid:173)
`mulation physical properties (e.g., collapse temperature) and process parameters
`(see below). The last criterion listed in Table 1 requires insight into the unique
`physicochemical properties of each therapeutic protein, which will be explained
`in more detail below. We will discuss in turn why each of these criteria is impor(cid:173)
`tant. Then we will present an explanation of how to design rationally a formula(cid:173)
`tion to meet these criteria.
`
`Table 1.
`Minimal Criteria for a Successful Lyophilized Protein Formulation
`
`1. Protein unfolding during freezing and drying is inhibited.
`2. The glass transition temperature of the product exceeds the planned storage temperature (e.g.,
`T, > 30°C).
`3. The water content is relatively low (e.g., 1% by mass).
`4. A strong, elegant cake structure is obtained (i.e., collapse and meltback are avoided).
`5. Steps are taken to minimize specific routes of protein chemical degradation (e.g., product vials
`are sealed under nitrogen to reduce the rate of methionine oxidation).
`
`
`
`
`

`

`112
`
`John F. Carpenter et al.
`
`Inhibition of Lyophilization-Induced Protein Unfolding
`
`The stresses of freezing and drying cause protein unfolding, and the fomm(cid:173)
`lation must be designed to inhibit unfolding at each step (Prestrelski et al.,
`1993a,b; Carpenter et al., 1993; Prestrelski et al., 1995; Constantino et al., 1995,
`1998; Griebenow and Klibanov, 1995; Allison et al., 1996, 1998, 1999, 2000;
`Chang et al., 1996b; Krielgaard et al., 1998a, 1999; Chen et al., 1999; Bell, 1999;
`Carrasquillo et al., 2000). Even if the formulation excipients and/or intrinsic ther(cid:173)
`modynamic stability of the protein prevent denaturation during freezing, unfold(cid:173)
`ing can arise during subsequent drying (Carpenter et al., 1993; Prestrelski et al.,
`1993b; Allison et al., 1998; Carrasquillo et al., 2000). Conversely, once a protein
`unfolds during freezing, it will not regain native structure during dehydration.
`For many proteins, unfolding during lyophilization leads to clinically
`unacceptable, non-native aggregates, even when samples are rehydrated immedi(cid:173)
`ately after lyophilization (Prestrelski et al., 1993a, 1995; Allison et al., 1996;
`Krielgaard et al., 1998a, 1999; Costantino et al., 1998). Aggregates are not nec(cid:173)
`essarily formed during freezing and drying. Rather, during rehydration refolding
`of structurally perturbed protein molecules competes with formation of non(cid:173)
`native protein aggregates (Prestrelski et al., 1993a). Aggregation can be mini(cid:173)
`mized by including stabilizing excipients (e.g., sucrose or trehalose) in the
`formulation to inhibit lyophilization-induced unfolding (Prestrelski et al., 1993a,
`1995; Allison et al., 1996; Krielgaard et al., 1998a, 1999; Costantino et al., 1998).
`Furthermore, fostering refolding during rehydration (e.g., with surfactants) can
`reduce aggregation (Chang et al., 1996c; Zhang et al., 1995, 1996).
`In addition to minimizing protein aggregation during lyophilization/rehy(cid:173)
`dration, maximizing retention of native protein structure in the dried solid is
`essential for optimizing long-term storage stability (Prestrelski et al., 1995; Chang
`et al., 1996b; Krielgaard et al., 1998a, 1999; Allison et al., 2000; Cleland et al.,
`2001). Both chemical and physical degradation in the dried solid can be acceler(cid:173)
`ated if protein unfolding is not inhibited during lyophilization. With chemical
`degradation, a non-native structure may provide an environment conducive to
`covalent modification of one or more residues. For example, exposure of a
`methionine, which is normally buried deep in the interior of the native protein,
`on the surface of an unfolded dried protein may foster oxidation. Increased levels
`of aggregates noted after storage and rehydration of unfolded proteins could be
`due to formation of non-native intermolecular contacts within the dried solid, per(cid:173)
`turbation of refolding during rehydration because of chemical degradation, and/ or
`other undefined processes.
`Infrared spectroscopy has been used routinely to compare the secondary
`structures of a protein in lyophilized formulations to that of the native protein in
`aqueous solution (Prestrelski et al., 1993a,b; Prestrelski et al., 1995; Dong et al.,
`
`
`
`
`

`

`Rational Design of Stable Lyophilized Protein Formulations
`
`113
`
`1995; Constantino et al., 1995, 1998; Griebenow and Klibanov, 1995; Allison et
`al., 1996, 1998, 1999, 2000; Chang et al., 1996b; Krielgaard et al., 1998a, 1999;
`Carpenter et al., 1998; Chen et al., 1999; Carrasquillo et al., 2000). This method
`should be considered essential in the development of stable lyophilized formula(cid:173)
`tions, because it allows one to assess rapidly the effectiveness of formulations at
`inhibiting protein unfolding. Technical details about how to employ infrared spec(cid:173)
`troscopy to design stable lyophilized protein formulations can be found in the
`papers cited above.
`
`Storage at Temperatures Below Formulation Glass .
`Transition Temperature
`
`In the dried powder, the protein is a component of an amorphous phase that
`includes amorphous excipients and water. If this glassy matrix is held below its
`characteristic glass transition temperature (T 8), the rate of diffusion-controlled
`reactions, including protein unfolding/aggregation and chemical degradation, are
`greatly reduced, relative to rates noted at temperatures >T8 (Roy et al., 1991;
`Franks, 1990; Franks et al., 1991; Pikal, 1994, 1999). T8 can be determined with
`differential scanning calorimetry (DSC) or other thermal scanning methods (Nail
`and Gatlin, 1993; Chang and Randall, 1992; Craig and Royall, 1998; Verdonck
`et al., 1999).
`Obtaining a formulation T8 in excess of the planned storage temperature
`(e.g., room temperature) is absolutely essential for optimal protein stability (e.g.,
`Franks et al., 1991; Pikal, 1994, 1999; Carpenter and Chang, 1996; Duddu and
`Dal Monte, 1997). The T8 of a given amorphous phase is dependent on the T8
`and mass percent of each component, including water (Angell, 1995; Franks
`et al., 1991; Levine and Slade, 1988, 1992; Pilcal, 1994, 1999). Compared to
`excipients, dried proteins have relatively high Tg's (e.g., >150°C; Angell, 1995).
`Thus, with all other factors being held constant, the formulation Tg varies directly
`with the mass fraction of protein. However, care must be taken that the mass frac(cid:173)
`tion of protein is not so high that there are not adequate levels of stabilizing excip(cid:173)
`ients to prevent protein unfolding during lyophilization (Cleland et al., 2001; and
`see below).
`Fortunately, sucrose and trehalose, which are the preferred excipients for
`inhibiting lyophilization-induced protein unfolding (see below), also provide a
`glassy matrix with acceptably high Tg values. For example, with water contents
`of 1% the T g for pure sucrose and trehalose are about 100 and 65°C, respectively
`(Crowe et al., 1998).
`It has now been documented with several proteins, that simply storing the
`formulation at temperature below Tg alone does not assure optimal stability. A
`
`
`
`
`

`

`114
`
`John F. Carpenter et al.
`
`native protein structure is also required. For example, proteins lyophilized in
`dextran alone are usually unfolded, but in a glassy matrix with a relatively high
`T 8 (e.g.,> 75° C). Yet they still degrade at relatively rapid rates compared to those
`for native protein molecules lyophilized with either sucrose or trehalose (Kriel(cid:173)
`gaard et al., 1998a, 1999; Lueckel et al., 1998; Allison et al., 2000; Yoshioka et
`al., 2000). On pharmaceutical time scales of several months of storage many
`degradative reactions are not coupled to the glass transition of a formulation. This
`is because on these times scales there is still significant molecular mobility, even
`at temperatures well below (e.g., more than 30° C) the T8 (Hancock et al., 1995;
`Duddu et al., 1997; Pikal, 1999; Yoshioka et al., 1999).
`
`The Water Content is Relatively Low
`
`Because of its very low T8 ( -135°C), wateris a potent plasticizerfor glasses;
`increasing water content in the dried formulation will greatly reduce T 8 • For
`example, increasing the water content of pure sucrose from 1 to about 3-4% (g
`H20/100 g dried powder) is sufficient to reduce the T 8 to below room temperature
`(Crowe et al., 1998). It is critical to achieve a suffiCiently low water level for a
`given formulation such that T8 exceeds the planned storage temperature. The
`lyophilization cycle dictates the initial water content (see reviews listed above).
`The most important parameter is the temperature for secondary drying, when the
`unfrozen water is desorbed (Pikal et al., 1990).
`Water can also be transferred to the product from the vial stoppers during
`storage (Pikal and Shah, 1992; DeGrazio and Flynn, 1992; Hora and Wolfe,
`1999). This effect can be dramatic. For example, let's consider a formulation con(cid:173)
`taining 10 mg of dried protein, 40 mg of sucrose and initial water content of 1%
`by weight. The total amorphous fraction containing protein and sucrose has 0.50
`mg of water. If l.Omg of water was transferred from the stopper to the product,
`the water content of this fraction would increase from 1% to 3.0%. This increase
`would be sufficient to lower the formulation T8 to below room temperature
`(Crowe et al., 1998). The risk of transfer of moisture from stoppers can be min(cid:173)
`imized by drying the stoppers before use, and, if acceptable for a given product,
`using stoppers coated with a material such as Teflon (see Hora and Wolfe, 1999).
`
`A Strong, Elegant Cake Structure is Obtained
`
`Often the most desired cake has strong, porous structure, without macro(cid:173)
`scopic collapse or meltback. This structure has a high surface area to volume
`
`
`
`
`

`

`Rational Design of Stable Lyophilized Protein Formulations
`
`115
`
`ratio, which aids in the rapid dissolution of product upon addition of water. A
`detailed account of how to obtain such a cake structure is beyond the scope of
`the current chapter, but is available in several previous reviews (see above). For
`the current purposes it is sufficient to focus on the impact of formulation com(cid:173)
`position on avoiding collapse or meltback. When a product is frozen, the protein
`and amorphous excipients (e.g., sucrose) are dispersed between ice crystals. and
`any excipient used as a crystalline bulking agent (e.g., glycine). To obtain an
`appropriate cake structure during lyophilization, the product temperature during
`primary drying, when the water in ice is sublimed, must be below the character(cid:173)
`istic collapse and eutectic melting temperatures of amorphous and crystalline
`solutes, respectively. Above the eutectic temperature, the melting of crystalline
`solutes leads to massive loss of porous structure and macroscopic dissolution of
`the frozen matrix into

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