throbber
Pharmaceutical
`Dosage Forms:
`Parenteral Medications
`Volume 1
`Second Edition, Revised and Expanded
`
`Edited by
`Kenneth E. Avis
`The University of Tennessee
`Memphis, Tennessee
`Herbert A. Lieberman
`HLH. Lieberman Associates,Inc.
`Consultant Services
`Livingston, New Jersey
`Leon Lachman
`Lachman Consultant Services
`Westbury, New York
`
`Marcel Dekker, Inc.
`
`New York « Basel « Hong Kong
`
`MAIA Exhibit 1028
`
`MAIA V. BRACCO
`
`IPR PETITION
`
`
`
`
`MAIA Exhibit 1028
`MAIA V. BRACCO
`IPR PETITION
`
`

`

`
`
`Library of Congress Cataloging - in - Publication Data
`
`Pharmaceutical dosage forms, parenteral medications / edited by
`Kenneth E. Avis, Herbert A. Lieberman, and Leon Lachman. -- 2nd ed.,
`rev. and expanded.
`p.-
`cm.
`Includes bibliographical references and index.
`ISBN 0-8247-8576-2 (v. 1: alk. paper)
`1. Parenteral solutions.
`2. Pharmaceutical technology.
`Kenneth E.
`II. Lieberman, Herbert A.
`III. Lachman, Leon.
`[DNLM: 1. Infusions, Parenteral.
`WB 354 P536]
`RS201.P37P48 1992
`615'. 19--dce20
`DNLM /DLC
`for Library of Congress
`
`I. Avis,
`
`2. Technology, Pharmaceutical.
`
`91 - 38063
`CIP
`
`This book is printed on acid-free paper.
`
`Copyright © 1992 by MARCEL DEKKER,INC. All Rights Reserved
`
`Neither this book nor any part may be reproduced or transmitted in any form
`or by any means, electronic or mechanical, including photocopying, micro-
`filming, and recording, or by any information storage and retrieval system,
`without permission in writing from the publisher.
`
`MARCEL DEKKER, INC.
`270 Madison Avenue, New York, New York 10016
`
`Current printing (last digit):
`0987654321
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`
`
`
`

`

`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`  ÿ 
`ÿ ÿ
ÿ ÿ
ÿ  ÿ
`ÿ
`ÿÿÿÿ
`
`7 P
`
`arenteral Products of Peptides
`and Proteins
`
`Yu-Chang John Wang
`California Biotechnology, Inc., Mountain View, California
`
`I.
`
`INTRODUCTION
`
`Scientific advances in molecular and cell biology have resulted in the develop-
`ment of two new biotechnologies. The first utilizes recombinant DNA to pro-
`duce protein products. Given the amino acid sequence of a protein, the gene
`responsible for producing such protein can be isolated, modified, and recom-
`bined with a plasmid DNA (Fig. 1). The modified plasmid can then be im-
`planted into a host cell. The recombinant DNA will be replicated and tran-
`seribed by the host cell to produce the specific protein in large quantities.
`Escherichia coli have been modified to produce growth hormone and growth
`factors, yeast to produce hepatitis B vaccine, and mammalian cells to produce
`erythropoietin. The advantages of such production are practically unlimited
`supplies of protein of high purity, and consistent quality in lot-to-lot produc-
`tion.
`The second technology involves fusion of antibody-producing B-lympho-
`cytes and myeloma (tumor) cells to form hybridomas (Fig. 2). Hybridomas
`grown in vivo in ascitic fluid or in vitro in tissue culture produce a single
`species of antibody, known as monoclonal antibody, which recognizes a spe-
`cific antigen.
`In contrast, conventional production methods in laboratory
`animals produce polyclonal antibodies that are comprised of a wide variety of
`antibodies with different affinity and antigenic specificities. Monoclonal anti-
`bodies, because of their unique properties of homogeneity, specificity, and
`affinity, can be used to:
`(1) isolate and purify proteins, e.g., Monoclat Fac-
`tor VIII:C is produced in this fashion; (2) neutralize a specific group of cells,
`e.g., Orthoclone OKT-3 neutralizes T-cells; (3) target™’specific cells or organs,
`e.g., antibodies to tumor antigens allow for tumor imaging or therapy by link-
`ing imaging radioactive isotopes or cytotoxic drugs to the antibodies.
`
`283
`
`
`
`
`

`

`284
`
`oe
`
`Donor DNA
`
`@
`

`
`@
`
`@
`
`Wang
`
`—_
`
`/
`Restriction
`gs
`
`enzymes
`
`Ligases (
`Recombinant
`
`ONA
`
`
`
`Plasmid DNA
`
`Hos? call
`
`Figure 1 Recombinant DNA for protein production. The technique of recom-
`bining genes from one species with those of another. Plasmid is an extrachro-
`mosomal, independently replicating small circular DNA molecule.
`(1) Restric-
`tion enzymes cut DNA at specific places. The donor DNA, represented by a
`heavy band, contains the information to produce the protein of interest.
`(2)
`Ligase fuses donor DNA with plasmid together.
`(3) Calcium is used to open
`cell wall or cell membrane to allow recombinant DNA to enter the host cell (bac-
`terium, yeast or mammalian cell).
`(4) Replication of the host cells and recom-
`binant DNA increase its number. Expression of the DNA produces desired
`protein.
`
`New products approved by the FDA between 1982 and 1989 derived from
`these two new biotechnologies are listed in Table 1.
`Although these two new biotechnologies have greatly expanded the capa-
`bility for producing large amounts of high quality proteins, not all new pro-
`tein products are produced in this way. Small proteins and peptides, such
`as calcitonin, may be produced by chemical synthesis. Human serum albumin
`is sourced from human blood, urokinase from urine, and streptokinase from
`fungi. Most of these proteins and peptides are formulated as injectable prod-
`ucts, although there are some exceptions. Cyclosporin is given per os. Oxy-
`tocin and desmopresin are available as nasal preparations. A topical ointment,
`Elase, contains two lytic enzymes, fibrinolysin and desoxyribonuclease, in
`lyophilized form. Wound-healing growth factors are being developed for use
`as topicals.
`Among the parenteral formulations, most of them are in aqueous solution
`or in freeze-dried form for reconstitution. However, one of the potent luteiniz-
`ing hormone release hormone (LHRH) analogues, leuprolide, and bovine somato-
`tropin have been developed as erodable microspheres for depot injection.
`
`Il. CHARACTERISTICS OF PROTEINS AND PEPTIDES
`
`A. Protein Structures
`
`Proteins and peptides are made of amino acids linked by peptide bonds. The
`sequence of these amino acids defines the protein's structure. The sequence
`of amino acids in human insulin is illustrated in Figure 3. A protein may also
`show a secondary structure, which is formed by either intrachain or inter-
`chain hydrogen bonds. These bonds may result in either an a-helix or §-sheet
`
`284
`
`284
`
`
`
`
`

`

`Parenteral Products of Peptides and Preteins
`
`285
`
`Antigen
`
`Ascitic fluid
`
`* c=
`se
`S
`Oo.
`Oo
`
`B-lymphocytes 4\_Fusion select are ib wr
`
`Ce or
`pe %
`0%
`vr
`Myeloma
`OQ
`Hybridoma
`Geacat
`calls
`
`antibody
`
`Fermenter
`
`(1) A mouse or rat is immunized
`Figure 2 Monoclonal antibody production.
`with a specific antigen. The spleen is removed and antibody-producing cells
`(B-lymphocytes) are isolated.
`(2) Transformed myeloma cells from another
`animal are isolated.
`(3) B-lymphocytes are fused with myeloma cells deficient
`in a particular enzyme, to form hybridoma cells.
`(4) Only the fusion product,
`hybridoma cells, can grow indefinitely in culture. The unfused myeloma cells
`and lymphocytes die. The hybridoma cells are cloned and ones expressing
`the desired antibody are selected to culture in large amounts. The antibody
`can be produced by these hybridomacells in either ascitic fluid or fermenta-
`tion tanks. The ascitic fluid or culture media is then processed to yield puri-
`fied monoclonal antibodies.
`
`structure. Figure 4 shows a peptide chain of a protein coiled to form an a-
`helix.
`The tertiary structure or conformation of a protein refers to the spatial,
`three- dimensional structure of the polypeptide chain. Given the primary and
`secondary structure, there are four kinds of interactions cooperating which
`may contribute to the tertiary structure of a protein. They are:
`(1) hydro-
`gen bonds between residues in adjacent loops of the chain, (2) ionic interac-
`tions (salt bridges) between oppositely charged residues, (3) hydrophobic
`interactions between the aliphatic or aromatic residues, or (4) disulfide link-
`ages (Fig. 5). Protein, in its native state, exists as a tight, compact folded
`structure. The folded structure of insulin is shown in Figure 6. When the
`tertiary or secondary structure is destroyed, the protein unfolds and is in
`its denatured state.
`If dimers, trimers, or other oligomers are formed, the arrangement of
`these subunits is a quaternary structure. The best-known example of an
`oligomeric protein is hemoglobin that contains four chains and no disulfide
`linkages. The quaternary structure is essential to its oxygen-carrying activ-
`ity.
`Insulin also forms dimers and, in the presence of zinc, hexamers. The
`role of the zinc insulin hexamers is to provide a storage form that is thermo-
`dynamically stable and more resistant to enzymatic degradation than the un-
`associated monomer.
`
`285
`
`285
`
`
`
`
`

`

`286
`
`Wang
`
`Table 1 Biotech Products Approved in the United States Between 1982
`and 1989
`
`Company
`
`Date approved
`
`Indication
`
`Drug
`
`Insulin
`
`Human growth
`hormone® (hGH)
`
`Interferon
`A2b (IFN)
`
`Lilly
`
`Genentech
`Lilly
`
`Schering
`Roche
`
`October 1982
`
`October 1985
`March 1987
`
`June 1986
`June 1986
`
`June 1986
`
`Diabetes
`
`Dwarfism
`
`Hairy cell
`leukemia
`
`Reverse kidney
`rejection
`
`Vaccine against
`hepatitis B
`
`Thrombolytic
`agent
`
`Muromonab-CD3
`
`Ortho
`
`Hepatitis B
`vaccine
`
`Tissue plasminogen
`activator (t-PA)
`
`Merck
`
`July 1986
`
`Genentech
`
`October 1987
`
`Erythropoietin (EPO)
`
`Amgen
`
`June 1989
`
`Anemia
`
`Hepatitis B vaccine
`
`SmithKline
`Biologics
`
`August 1989
`
`Vaccine against
`hepatitis B
`
`“Growth hormone from Genentech differs from Lilly's product by a single
`methionine at the N-terminus.
`binterferons from Schering and Roche differ by one amino acid.
`
`In some proteins, there are carbohydrates attached to specific amino acid
`residues to form glycoproteins. For example, erythropoietin is a glycoprotein
`hormone with a molecular weight of 44 kilodaltons, having 40% of its weight
`accounted for by sialic acid, an amino sugar. Carbohydrate residues stabilize
`the tertiary structure and may influence receptor binding, bioactivity, and
`pharmacokinetics of the protein in vivo.
`
`B.
`
`Isoelectric Point
`
`In the course of a potentiometric titration of a protein there is a pH at which
`the mean charge on the protein is zero, This pH is termed the isoelectric
`point, pl. Table 2 provides examples of pIs for a number of therapeutic pro-
`teins and peptides. The pI may be approximately calculated from the amino
`acid composition data, i.e., pl = (pK; + pKg + pKg.
`.
`. + pK,,)/n for n ioniz-
`able groups. However, because the dielectric constant in the immediate vicin-
`ity of an ionizable group depends on protein structure and, because hydrogen
`bonding may alter dissociation constants (Kgs), the true pI can differ signifi-
`cantly from the calculated one.
`Just like pKgs of small organic compounds, the pI of a protein plays an
`important role in solubility.
`In general, near the pH of the isoelectric point,
`protein solubility is at its minimum. This trend is similar to the low solubility
`exhibited by a zwitterion compound.
`
`286
`
`286
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`287
`
`s—-s
`
`
`pooozys(S&1oraGLIAL24d4dATOBay|nqo[<t5shoTeA|NeTIALeTETYOTS|TEANOTSTHzegTDShQNe]STHUTO|USYTPAaqa°aN
`OfSt0zSTorST
`
`
`
`moo@Joseazoued94}Worpe}B[OSTSUT[NSUTysnoyyly‘eseyxUTepyMsrpTeusezuyueseysuTeyoay}jo
`
`
`
`
`
`ey}pue[TZUMUTeYydVYey}‘SUTeYyoOM}seyUTMsSUL“UPNSUTuewnyjosduenbespoeoulwy¢eanbi4
`
`
`
`
`auopuB‘SasBYUT[-SSOAOapIJ[NSIpOM}AqpeyoeuuodaresureyoOM}CYL“SpeOUTUTEOFUFIMureyo
`
`
`
`
`uedUT[NSUTUBUINY,=*“pexoqoie‘satoedsSuourejuelseaul‘*e*t‘senp{sedploeourwepaaresuooATySTYy
`
`
`
`
`
`
`-suei}orjewAzueueAqUTNsUryaodwoay10VNJUeUIqWODeTYIMyseak10909"gyAQpeonpordaq
`
`
`auL‘UT[NSUTUBUINYYIM[eoT}UeprjoueBAou}‘sjuerjedotjeqerpjoJUSUI}ee1}UTpesnodB31d@20
`
`
`
`
`
`
`
`
`ay}Sutwaoysny}‘O&goutueyeay}LoyeuTUOeTY}BseinyisqnsApeaTOeTesYOTUMsseoo0rduorjepyded
`
`
`[wooousyS49241)ATT795eento[Tea®TT4T9¢HNusyngnayUTDryney19s[s49|
`/ss/|IzscIoTs¢
`
`
`
`
`
`
`
`*sernoe[owuT;NsuUT-uBeUNY
`
`287
`
`287
`
`
`
`
`
`

`

`288
`
`Wang
`
`Figure 4 Peptide chain of a protein coiled to form an a-helix. Configuration
`of the helix is maintained by hydrogen bonds shown as vertical dotted lines.
`
`C. Degradation Through Covalent Bonds
`
`The degradation of proteins and peptides can be divided into two main cate-
`gories:
`those that involve a covalent bond and those involving a conforma-
`tional change. The latter process is often referred to as denaturation.
`Numerous types of chemical reactions leading to modification of the co-
`valent structure are possible in a protein. The following describes only those
`that are likely to be seen in a protein or peptide formulation.
`
`Peptide Fragmentation
`
`The peptide bond (RNH—CO—R) is much moreresistant to hydrolysis than is
`the ester linkage (R—O—CO—R) and peptide bonds are considered stable un-
`
`288
`
`288
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`289
`
`Disulfide
`Bond
`
`
`
`
`fonic
`lnteraction
`
`Figure 5 Interactions that stabilize folded conformation.
`
`less hydrolysis is assisted by a neighboring group. The formulation factor
`that most influences the hydrolytic rate is solution pH. The rate of hydroly-
`sis is in direct proportion to the activity of hydronium, or hydroxide ions,
`when in acidic or alkaline pHs, respectively. A rate-pH profile for hydrolysis
`of tetraglycine is shown in Figure 7. The reaction minimum is in the region of
`PH 3to 5 [2]. Published examples of stability studies on therapeutic peptides
`are: degradation of nafarelin [3], thermal stability and degradation mecha-
`nism of secretin [4], acid hydrolysis of captopril, a quasidipeptide [5], and
`heat stability of urokinase [6].
`Many peptides are stable enough to formulate as a ready-to-use parenteral
`formulation. For instance, D-Trp-LHRH was stable at 60°C for 5 days and
`seven cycles of freezing and thawing; gramicidin, although used only for derma-
`tological indications when dissolved in glycols, can be autoclaved.
`Information
`available on package inserts also shows it is possible to have good long-term
`stability of peptides. Leuprolide injection has a shelf-life of at least 2 years
`under refrigeration and 3 months at room temperature. LH-RH injection (Rele-
`fact) does not even require storage under refrigeration. Oxytocin injection
`was reported to be stable at room temperature for 5 years [7]. Protirelin, a
`tripeptide (PyrGlu-His-Pro), is stable for 20 hr at 80°C at both pH 3.3 and
`pH 6 [8].
`Certain amino acids form the weak link of the chain. The bond between
`aspartic acid and proline or tyrosine is sensitive to acid hydrolysis [9]. The
`resultant products are peptides with aspartic acid at the C-terminus. The C-
`terminal peptide bond adjacent to serine is also a reactive one due to the neigh-
`boring group effect of the alcohol on serine [10].
`
`289
`
`289
`
`
`
`
`

`

`290
`
`Wang
`
`Figure 6 Folded three-dimensional structure of insulin. The A-chain is ar-
`ranged between the two terminal arms of the B-chain and lies on top of the
`central a-helix (B9-Bj9). The hydrophobic regions are responsible for the
`association of monomer into dimers and hexamers.
`(Adapted from Ref. 1.)
`
`Table 2 pls of Selected Proteins
`
`
`
` Proteins pI
`
`Pepsin
`
`Erythropoietin
`
`Serum albumin
`
`Growth hormone
`
`Insulin
`
`Interleukin- 2
`
`Calcitonin (salmon)
`
`Chymotrypsin
`
`Basie fibroblast growth factor
`
`Interferon- y
`
`Lysozyme
`
`Nafarelin
`
`1.0
`
`3.5-4.0
`
`4.8
`
`5.0
`
`5.4
`
`7.8
`
`7.8
`
`9.1
`
`9,8
`
`10.0
`
`11.0
`
`11.3
`
`
`
`Protamines 12.0
`
`290
`
`290
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`291
`
`=2
`
`
`
`LogKopa(hr”)
`
`pH
`
`Figure 7 Rate-pH profile for the hydrolysis of tetraglycine. Peptide solu-
`tions containing phosphate buffer, 0.03 M, and sodium chloride, 0.1M, were
`incubated at 60°C. Samples were analyzed by HPLC assay. At pHs 1 to 3,
`the slope is -1, indicating an acid catalyzed reaction.
`In the pH region 4 to
`7, the reaction is hydroxide ion catalyzed. The plateau at pH 8 is caused by
`change in ionization state, zwitter ion to negatively charged ion, which is less
`susceptible to hydroxide-ion attack [2].
`
`Deamidation
`
`Deamidation refers to the removal of ammonia from the amide (RCONHg) moiety,
`whetherit is at the end of a C-terminus or at asparagine (Asn) or glutamine
`(Gln) residues. Stability studies of insulin [11], ACTH [12], human growth
`hormone [13], and RS-26306 [10], an LH-RH analogue, have shown this route
`to be the major factor resulting in instability of these proteins and peptides
`in aqueous solution.
`In acidic media, the peptides deamidate by direct hydrolysis. Thus, an
`Asn or Gln residue yields an Asp or Glu peptide, respectively. The neighbor-
`ing amino acid does not affect the deamidation rate. Hydrolytic mechanisms
`in neutral or alkaline pHs are more complex, however. Under these condi-
`tions, the side chain carbonyl group on the Asn or Gln residue attaches to
`the nitrogen atom on the peptide backbone to form a cyclic imide intermediate.
`Depending on which bond in the cyclic imide breaks (Fig. 8), the reaction
`product can be:
`(1) the des-amido peptide, (2) the isopeptide, or (3) D-
`isomers. The formation of isopeptide is sometimes referred to as transpeptida-
`tion because an extra methylene group is inserted to the peptide backbone.
`At neutral to alkaline pHs, the rate of deamidation is significantly affected by
`the size of the amino acid on the C-terminal side of the Asn or Gin residue.
`In general, Asn is more labile than Gln, and is most labile when adjacent to
`glycine, which is the least obstructive to the formation of a cyclic imide.
`Deamidation rate-pH profile is V-shaped [14], usually with a minimal rate
`at a pH of about 4 to 5 (Fig. 9).
`In a number of synthetic peptides the half-
`
`291
`
`291
`
`
`
`
`

`

`292
`
`Wang
`
`1
`-C-O° RO
`CHa
`|
`Jirarte
`|
`R'-NH-C—<”
` Deamidation
`§ for Asn & Gin
`
`0
`
`0
`

`
`0
`
`=
`
`”
`a” eo
`1
`ott
`R'-NH—-CH-C-NH-CH-C ~
`i
`NH,
`a i
`a
`CHa yy Yo
`.
`Tl
`fe
`Che
`|
`N—CH—C~ aga
`NH-CH-C ~
`R'-NH-C__*b
`R'-NH C~¢ 09
`Hc §
`6 Transpeptidation
`oO
`
`a
`
`c
`
`Imide Formation
`
`oO
`R
`me
`&
`ey T.
`UH N-CH-C~
`R'=NH-CW °7 Fracemization
`
`il

`
`Figure 8 Reaction pathways in deamidation of asparagine. Deamidation of
`asparagine or glutamine goes through an intermediate, a cyclic imide. Three
`routes of degradation can take place, which leads to products of deamidation
`(removal of the amino group), transpeptidation (insertion of an extra methy-
`lene group), and racemization (D aspartic or glutamic acids).
`
`Logky(hr)
`
`ro
`
`0
`
`2
`
`4
`
`8
`
`10
`
`12
`
`6
`pH
`
`Figure 9 Deamidation rate-pH profile of an active segment of adrenocortico-
`tropic hormone, Val-Tyr-Pro-Asn-Gly-Ala, in aqueous solution at 37°C and
`ionic strength of 0.5. The apparent rate is followed by the disappearance of
`the parent peak in HPLC.
`(From Ref. 14.)
`
`292
`
`292
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`293
`
`lives of deamidation reactions of Asns at 37°C in pH 7.2 phosphate buffer
`range from 2 days to nine years [15]. Not all Asns are equally labile; those
`buried within the interior portion of a protein are inaccessible to water and
`thus less reactive.
`In a large protein, secondary and tertiary structures
`play an important role in determining the site and the rate of deamidation.
`There are three asparagines in insulin (Fig. 3), the monodesamido-(A21)-
`insulin is the prevailing degradant formed in acid solution.
`In neutral solu-
`tion, deamidation is slow at 5°C. However, when accelerated at higher stor-
`age temperatures, deamidation of Asn at the B3 position can be detected [11].
`Figure 10 shows deamidation of insulin in neutral formulations at three differ-
`ent temperatures. With growth hormone, there are nine asparagines among
`191 amino acids, and deamidation oceurs primarily at the Asn-149 position [13].
`
`Oxidation of Cysteine
`
`Under neutral or basic conditions, the free thiol (—SH) group of a cysteine
`is the most reactive moiety of all amino acid components. The disulfide
`(—S—S—) bond formed from the oxidation of two thiol groups results in sig-
`nificant changes in conformation both intramolecularly and intermolecularly.
`Oxidation of the thiol group is promoted at both neutral and basic pH.
`The rate-pH profile for captopril, a quasi dipeptide, showed an increase in
`oxidation rate, starting at pH 5 (Fig. 11). This reaction can be effectively
`retarded by the addition of a metal chelating agent such as EDTA [5].
`A strategy to stabilize proteins that have reactive thiol groups is to re-
`place the cysteine with serine. Serine, a stable amino acid with a hydroxy
`(—OH) group, mimicks the size and polarity of cysteine.
`In many cases,
`these serine mutants retain full biological activities.
`
`Oxidation of Methionine
`
`The methiol (—S—CHg) moiety on methionine is susceptible to oxidation to
`form methylsulfoxide (—SO—CH3) derivatives. Of the three methionines in
`human growth hormone, Met-125 is most reactive, Met-14 is less so and Met-
`170 is not reactive at all [16]. The reactive methionine is likely to be the
`one exposed on a protein surface, and the unreactive one buried within the
`core. Air in the headspace of formulated and freeze-dried growth hormone
`can cause 40% of the growth hormone molecules to be oxidized during a 6 month
`storage period [17].
`
`Disulfide Exchange
`
`Disulfide exchange takes place when a cystine (disulfide) bond is reduced
`to two cysteines; one of them then reacts with another cysteine to form a new
`disulfide (Fig. 12). The reaction is base (hydroxide ion) catalyzed and pro-
`moted by mercaptoethanol, which is sometimes used as an antioxidant. The
`reaction is concentration-dependent, and oligomers are frequently formed as
`a result of disulfide scrambling of bonds between peptide chains.
`Improper
`linkages of disulfide bonds were responsible for a reduction in biological ac-
`tivity of interleukin-2 (IL-2) (Fig. 12). There are three cysteines in IL-2
`at positions 58, 105, and 125, The native protein forms a disulfide linkage
`between the two cysteines at 58 and 105. Cleavage of this disulfide in IL-2
`and the subsequent formation of two less-active isomers with disulfide bonds
`
`293
`
`293
`
`
`
`
`

`

`Wang
`
`294
`
`%deamidation
`
`Years
`
`Figure 10 Deamidation of insulin during storage of Actrapid MC (BP-formula-
`tion) at different temperatures. Each point represents the mean of analyses
`of 4-6 different batches. The desamido insulin content was determined by
`basic disc electrophoresis followed by densitometric scanning of the stained
`gels.
`(From Ref. 11.)
`
`at incorrect positions are promoted by high pH and copper ions [18]. The
`number of disulfide bonds in therapeutic proteins can be shown in the follow-
`ing examples:
`IL-2:1; salmon calcitonin:1; interferon-a:2; human growth
`hormone: 2; insulin: 3; urokinase: 12;
`t-plasminogen activator:17; albumin: 17.
`
`50
`
`nm
`2 30
`
`40
`
`
`7 7w a2
`
`2a=
`
`10
`
`q
`N-C-CH-CH,-SH ——= R-S-S-R
`CH,
`COOH
`
`Figure 11 Oxidation of the thiol moiety in captopril. Reaction rates at 50°C
`is followed by a peak on the HPLC chromatogram. All solutions contain di-
`sodium edetate. Without the chelating agent, reaction rates are much faster.
`(From Ref. 5.)
`
`294
`
`294
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`295
`
`Native Interleukin - 2
`
`CoSs
`
`Denoturing
`Conditions
`
`ass =Z \AlkalinepH
`
`ae
`08252 25
`SH
`
`sa)
`
`05)-SH
`ssqes
`
`Less active forms
`
`Figure 12 Schematic representation of the various disulfide-linked isomers of
`interleukin-2 [18]. Under denaturing conditions, i.e., high temperature or
`alkaline pH, a free sulfhydryl group forms a disulfide bond with the sulfhy-
`dryl group from an existing disulfide. The results are proteins with incorrect
`disulfide linkages and "non-native" conformation.
`
`With N-disulfide bonds, there exist (2N)!/2N x N! possible isomers. Cau-
`tion must be exercised to preserve the correct disulfide linkage throughout
`the production process for therapeutic proteins.
`
`Racemization
`
`The racemization reaction is catalyzed by both acid and base. Racemization
`of peptides and proteins results in the formation of diastereomers. As an
`example, racemization of the serine residue in a gonadotropin-releasing hor-
`mone analogue was one of the main degradation reactions when treated with
`0.1 N NaOH at 5°C for 48 hr [19]. Racemization under basic conditions is
`thought to proceed by abstraction of the c-proton from an amino acid in a
`peptide to give a negatively charged planar carbanion. A proton can then be
`returned to this optically inactive intermediate, thus producing a mixture of
`D- and L-enantiomers for the individual amino acid (Fig. 13). Since a pep-
`tide is composed of multiple chiral centers, the product formed is a diastereo-
`mer. Racemization is biologically significant since a peptide comprised of D-
`amino acids is generally metabolized much slower than a naturally occurring
`peptide made of only L-amino acids. For this reason, many new synthetic
`peptides, agonists or antagonists, incorporate D-amino acids. A pH depend-
`ency on racemization was demonstrated in an aqueous degradation study of a
`decapeptide, RS-26306 [10]. At neutral and alkaline pHs, racemization con-
`tributed to more degradation than deamidation (Fig. 14).
`
`295
`
`295
`
`
`
`
`

`

`296
`
`Wang
`
`CHR O
`I
`ll
`
`CHR O
`|
`ll
`
`H
`I
`
`QO
`ll
`
`eioer =_— *NH-C——C- —— SeeS
`ne &
`CHR
`OH
`
`Figure 13 Racemization of an amino acid resulting in a mixture of enantiomers.
`
`Maillard Reaction
`
`In the Maillard reaction, the carbonyl group (RCH=0) from glucose can react
`with the free amino group (e.g., R'NHg in lysine) in a peptide to form a
`Schiff base (RCH=NR'). This reaction is acid catalyzed. During pasteuriza-
`tion (60°C, 10 hr), the loss of antithrombin III biological activity seen in the
`presence of reducing sugars, especially glucose, was attributed to this reac-
`tion [20]. An alternative assessment of poor blood-glucose control in diabetic
`patients is the measurement of glycosylated hemoglobin, which is the product
`of the Maillard reaction. The hospital pharmacist should thus be concerned
`about storing protein in dextrose solution for a prolonged period of time.
`
`oe
`
`io7>
`
`io~*
` Epimerization
`
`pH
`
`Figure 14 Rate-pH profiles for degradation of RS- 26306, N-acetyl-3-(2-naph-
`thyl)-D-Ala- 4- Chloro-D-Phe- 3-(3-pyridinyl)-D-Ala-Ser-Tyr-N, N-diethyl-D-
`Arg-Leu-N, N-diethyl-Arg-Pro-D-Alanylamide, a decapeptide [10].
`
`296
`
`296
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`297
`
`Dimerization and Polymerization
`
`Insulin forms a small amount (about 1%) of covalent dimer and polymer during
`2 years storage at 4°C. Production of these species increases dramatically
`with increasing temperature. The dimerization is mainly due to a reaction be-
`tween an N-terminal amino group in one insulin with a carboxamide group of
`a glutamine or an asparagine in another insulin molecule [11].
`The covalent bond reactions aforementioned are examples of key reactions
`that may cause protein or peptide instability during storage. A complex pro-
`tein may undergo many reactions simultaneously.
`
`D. Denaturation
`
`Specific conformation is required for proteins to exert physiological and phar-
`macological activities. Denaturation is a process of altering protein conforma-
`tion. The denaturation process is illustrated in Figure 15 [21]. The folded
`structure (N) unwinds, passes through a critical activated state (A*), to an
`incompletely disordered conformation (ID). Finally, some of the ID forms
`uncoil to become random boils (RC). Heat, extreme pHs, organic solvents,
`high salt concentration, lyophilization, or mechanical stress can denature pro-
`teins.
`
`ID
`
`RC
`Level
`
`Energy
`
`Reaction Coordinate
`
`Figure 15 Schematic illustration of reversible conformational transitions of
`a-lactalbumin [21]. N, Native conformation; A*, critical activated state; ID,
`incompletely disordered conformation; and RC, random coil (fully denatured
`state).
`
`297
`
`297
`
`
`
`
`

`

`298
`
`Wang
`
`Protein conformation refers to the specific tertiary structure, which is
`determined by the primary and secondary structures and the disulfide bonds,
`and is held together by three forces: hydrogen bonding, salt bridges, and
`hydrophobic interactions (Fig. 5).
`Hydrogen bonds are the most important in determining overall protein
`conformation, since they are the major forces that stabilize the secondary
`a-helices and §-sheets, as well as the overall folded structure. Water, the
`ubiquitous medium for most proteins, contributes to this hydrogen bonding.
`Thus, to avoid denaturation in protein during a freeze-drying process, a
`small amount of residual moisture is critical for recovering biological activity
`of the reconstituted protein. Cosolvents such as ethanol and acetone, and
`chaotropic agents such as urea and guanidinium chloride disrupt the hydro-
`gen bonds thus readily denature proteins.
`The ionic side-chains of aspartic acid, glutamic acid, lysine, arginine,
`and histidine, normally found on the surface of the protein, contribute to
`the stability of the native conformation by forming salt bridges. The pH of
`the solvent will determine the charge of the side-chains on these amino acids
`and the extent of ionic bonding. Thus, an extreme pH shift can disrupt these
`salt bridges and lead to denaturation, Further, organic solvents weaken the
`streangth of salt bridges, thus, inappropriate exposure to organic solvent
`can also result in denaturation.
`Because hydrophobic side-chains, i.e., phenyl and hydrocarbon chains,
`are usually tucked inside the protein's globular structure, significant stabil-
`izing effects result from their hydrophobic interactions. This interaction,
`too, is sensitive to the effects of solvents. Disruption of hydrophobic inter-
`actions is often considered the mechanism of denaturation by surfactants,
`heat, mechanical stress, or storage. When a protein molecule unfolds under
`thermal stress, and before it refolds to its natural conformation, hydrophobic
`interaction can result in one of two different types of unnatural conformations.
`In a concentrated protein solution, the hydrophobic groups may interact be-
`tween molecules, resulting in protein aggregation.
`In dilute solutions of pro-
`tein, however, intramolecular interactions are much more likely, and may re-
`sult in a conformation different from the native one. Such unnatural conforma-
`tions may show partial or complete loss of biological activity.
`
`E. Analytical Methods
`
`For protein products a battery of tests are required to assure identity, purity,
`potency, and stability of the formulation. Due to complexity of proteins, as
`of yet no chemical or physical assay can substitute for a bioassay for assess-
`ing the potency. There are in vitro and in vivo bioassays. The in vitro bio-
`assay monitors the response of cells or excised tissue to the stimulation of
`hormones, growth factors, or antibodies. For example, the activity of IL-2
`is determined by measuring [°H]-thymidine incorporated into an IL-2 depend-
`ent murine T-cell line. The in vivo bioassay monitors the pharmacological
`responses of animals to the proteins. For example, the activities of insulin
`and glucagon are determined by measuring post-injection blood sugar in rab-
`bits and cats, respectively. Often, in vitro and in vivo bioassays are not
`sensitive enough to detect subtle changes in protein characteristics. Thus,
`consistency in product performance relies heavily on validated manufacturing
`
`298
`
`298
`
`
`
`
`

`

`Parenteral Products of Peptides and Proteins
`
`299
`
`processes as well as end product testing. When the manufacturing process
`or formulation is significantly changed, the only assured way to prove bio-
`equivalency is to conduct clinical studies in patients.
`The multiplicity of degradation routes for proteins requires combined in-
`formation from various assays to assess the stability of the product. The
`following sections describe common analytical methods employed in protein
`formulations.
`
`UV Spectrometry
`Any protein with at least one tryptophan residue can be detected, at about
`the 0.1 mg/ml level, by ultraviolet (UV) spectroscopy at 270 to 280 nm.
`Phenylalanine and tyrosine contribute about one-sixth the molar absorptivity
`to that of tryptophan. One may use UV for batching assay because of its
`precision, reproducibility, and simplicity. The absorptivity can be deter-
`mined from literature values or derived from the absorbance and mass ob-
`tained from amino-acid composition. For example, the generally accepted value
`for the absorbance at 275 nm of a 1 mg/ml solution of immune globulin is 1.5.
`The UV method can be a rapid assay for in-process quality control. Also,
`because protein aggregates scatter UV light with increased absorbance over
`the range 210 to 350 nm, UV spectrometry can be used to monitor protein ag-
`gregation.
`
`Proteins Assay
`Bradford, Lowry and bicinchoninic acid (BCA) assays are three commonly
`employed colorimetric assays for determination of protein content.
`The Bradford assay is based on a change in the absorption maximum,
`from 465 to 595 nm, of an aromatic sulfonate dye, Coomassie Brilliant Blue
`G-250, in the presence of protein.
`Biuret, with a structure of NHgCONHCONHg, and peptides,

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