throbber
METHODS IN AMOLECULARBIQLOCY
`
`VölumeZ98
`
`Pepide
`Syuliesis
`andpplicatians
`
`Edied by
`ohu Howl
`
`KHUMANAWBNASS
`
`MPI EXHIBIT 1058 PAGE 1
`
`

`

`Peptide Synthesis.and Applications
`
`MPI EXHIBIT 1058 PAGE 2
`
`

`

`METHODS IN MOLECULAR BIOLOGYt#
`
`Peptide Synthesis
`and Applications
`
`Edited by
`John Howl
`Research Institute in Healthcare Science,
`School of Applied Sciences, University of Wolverhampton,
`Wolverhampton, UK
`
`HUMANA PRESS
`
`TOTOWA, NEWJERSEY
`
`MPI EXHIBIT 1058 PAGE 3
`
`

`

`O 2005 Humana Press Inc.
`999 Riverview Drive, Suite 208
`Totowa, New Jersey 07512
`
`www.humanapress.com
`
`All rightsreserved. No part of this book may be reproduced, stored in a retrieval system or
`
`transmite
`
`anyformor byanymeans,electronic,mechanical, photocopying, microfilming,recordinetrademarkeofThe
`eijge
`without witten permission from the Publisher. Methods in Molecular BiologyTM is atrod
`
`Humana Press Inc.
`
`All papers,comments, opinions, conclusions, or
`necessarilyreflect the views of the publisher.
`
`recommendations are those of theauthor(e)
`,anddonm
`
`Thispublicationisprintedonacid-freepaper. o
`ANSI Z39.48-1984 (American Standards Institute)
`
`Permanenceof Paper for Printed ILibrary Materials.
`
`Production Editor: C. Tirpak
`Cover design by Patricia F. Cleary
`
`information about other Humana titles,contlg
`For additional copies, pricing for bulk purchases, and/or
`Humana at the above address or at any of the following numbers: Tel.: 973-256-1699; Fax:973-256-841:
`E-mail: humana@humanapr.com; or visit our Website: www.humanapress.com
`
`Photocopy Authorization Policy:
`Authorization to photocopy items for internal or personal use, or the internal or personal useofspecife
`clients, is granted by Humana Press Inc., provided that the base fee of US $30.00 per copy ispaiddireciy
`to the Copyright Clearance Center at 222 Rosewood Drive, Danvers, MA 01923. For thoseorganizations
`that have been granted a photocopy license from the CCC, a separate system of payment hasbeenarranged
`and is acceptable to Humana Press Inc. The fee code for users of the Transactional ReportingServiceis
`(1-58829-317-3/05 $30.00 ).
`
`PrintedintheUnitedStatesof America. 10 9
`
`87654321
`
`elSBN:1-59259-877-3
`
`Library ofCongress Cataloging-in-Publication Data
`
`Peptidesynthesis and applications / edited by John Howl.
`
`P. ; cm. -
`
`(Methods in molecular biology; 298)
`
`Includes bibliographical references and index.
`
`ISBN 1-58829-317-3 (alk. paper)
`
`1.
`
`Peptides-Synthesis--Laboratory manuals.
`
`(DNLM: 1. Peptide Synthesis--Laboratory Manuals. QU 25 P4246 2005]
`
`I.
`
`Howl, John. II. Series: Methods in molecular biology (Clifton, N.J.) ; v.
`298.
`
`QD431.25.S93P47 2005
`
`547.756--dc22
`
`2004020037
`
`MPI EXHIBIT 1058 PAGE 4
`
`

`

`Contents
`
`Preface
`
`Contributors
`
`PART I: COMMON STRATEGIES
`
`1
`
`Fundamentals of Modern Peptide Synthesis
`Muriel Amblard, Jean-Alain Fehrentz, Jean Martinez,
`and Gilles Subra ..
`2 Chimerism: A Strategy to Expand the Utility
`and Applications of Peptides
`JohnHowl .....
`
`PART I1: SYNTHETIC METHODOLOGIES AND APPLICATIONS
`
`3 Modification of Peptides and Other Drugs Using
`Lipoamino Acids and Sugars
`Joanne T. Blanchfield and Istvan Toth..
`
`4 Synthesis of Linear, Branched, and Cyclic Peptide Chimera
`Gábor Mezö and Ferenc Hudecz...
`
`6
`
`5 Synthesis of Cell-Penetrating Peptides for Cargo Delivery
`Margus Pooga and Ülo Langel....
`Incorporation of the Phosphotyrosyl Mimetic
`4(Phosphonodifluoromethylphenylalanine (F,Pmp)
`Into Signal Transduction-Directed Peptides
`Zhu-Jun Yao, Kyeong Lee, and Terrence R. Burke, Jr.
`
`7 Expressed Protein Ligation for Protein Semisynthesis
`and Engineering
`Zuzana Machova and Annette G. Beck-Sickinger....
`
`8 Cellular Delivery of Peptide Nucleic Acid
`by Cell-Penetrating Peptides
`Kalle Kilk and Ülo Langel...
`9 Quenched Fluorescent Substrate-Based Peptidase Assays
`Rebecca A. Lew, Nathalie Tochon-Danguy,
`Catherine A. Hamilton, Karen M. Stewart,
`Marie-Isabel Aguilar, and A. lan Smith .....
`
`vii
`
`.V
`...ix
`
`ooooness3
`
`****.*.... 25
`
`45
`
`..
`
`..... 63
`
`*. 77
`
`91
`
`eeeeeeeeo
`
`105
`
`131
`
`143
`
`MPI EXHIBIT 1058 PAGE 5
`
`

`

`Vii
`
`Content
`
`10 A Convenient Method for the Synthesis of Cyclic Peptide I :A.
`braries
`Gregory T. Bourne, Jonathon L. Nielson, Justin F. Coughlan.
`Paul Darwen, Marc R. Campitelli, Douglas A. Horton,
`Andreas Rhümann, Stephen G. Love, Tran I. Tran,
`and Mark L. Smythe.
`
`0000eeeeesesess,
`*****1$1
`
`11
`
`High-Throughput Peptide Synthesis
`Michal Lebl and John Hachmann
`
`12 Backbone Amide Linker Strategies for the Solid-Phase Synthesis
`of C-Terminal Modified Peptides
`Jordi Alsina, Steven A. Kates, George Barany,
`andFernando Albericio...
`
`eeeee
`
`19%
`
`13 Synthesis of Peptide Bioconjugates
`FerencHudecz ....
`
`PART III: PRACTICALGUIDES
`
`eeeeeeeeeeeses,
`
`20
`
`14 Protein ldentification by Mas Spectrometric Analyses ofPeptides
`Ashley Martin ...
`15 Manual Solid-Phase Synthesis of Glutathione Analogs:
`A Laboratory-Based Short Course
`UrselSoomets, Mihkel Zilmer, and Ülo Langel...
`Index...
`
`•ee0eeeeeesssss
`
`22
`
`24
`
`250
`
`MPI EXHIBIT 1058 PAGE 6
`
`

`

`1 F
`
`undamentals of Modern Peptide Synthesis
`
`Muriel Amblard, Jean-Alain Fehrentz, Jean Martinez, and Gilles Subra
`
`Summary
`The purpose of this chapter is to delineate strategic considerations and provide
`practical procedures to enable non-experts to synthesize peptides with a reasona-
`ble chance of success. This chapter focuses on Fmoc chemistry, which is now the
`most commonly employed strategy for solid phase peptide synthesis (SPPS). Proto-
`cols for the synthesis of
`fully deprotected peptides are presented, together with a
`review of linkers and supports currently employed for SPPS. The principles and the
`different steps of SPPS (anchoring. deprotection, coupling reaction, and cleavage)
`are all discussed, along with their possible side reactions.
`
`Key Words: Solid phase peptide synthesis; side reaction coupling; anchoring;
`deprotection; cleavage;
`linker.
`
`1.
`
`Introduction
`
`Nowadays, "peptide synthesis" includes a large range of techniques and pro-
`cedures that enable the preparation of materials ranging from small peptides to
`large proteins. The pioneering work of Bruce Merrifield (1), which introduced
`solid phase peptide synthesis (SPPS), dramatically changed the strategy of pep-
`tide synthesis and simplified the tedious and demanding steps of purification
`associated with solution phase synthesis. Moreover, Merrifield's SPPS also per-
`mitted the development of automation and the extensive range of robotic instru-
`mentation now available. After defining a synthesis strategy and programming
`the amino acid sequence of peptides, machines can automatically perform all
`the synthesis steps required to prepare multiple peptide samples. SPPS has now
`become the method of choice to produce peptides, though solution phase syn-
`thesis can still be useful
`for large-scale production of a given peptide.
`
`From: Methods in Molecular Biology, vol. 298: Peptide Synthesis and Applications
`Edited by: ). Howl O Humana Press Inc., Totowa, NJ
`
`3
`
`MPI EXHIBIT 1058 PAGE 7
`
`

`

`4
`
`Amblard etal.
`
`frit
`
`to vacuum
`
`Fig. 1. Basic equipment
`
`for SPPS.
`
`2. Materials
`
`(Fig. 1).
`1. Reaction vessel
`2. Polytetrafluoroethylene (PTFE) stick (15 cm length, 0.6-0.8 cmdiameter).
`3. Rotor.
`4. Filtration flask.
`5. Porous frit.
`6. Lyophilizer.
`7. HPLC equipped with reverse phase Cis column.
`8. pH-Indicating paper.
`9. Solvents
`(N,N-dimethylformamide
`(DCM)
`in wash bottles.
`(DIPEA).
`10. Diisopropylethylamide
`11. Piperidine solution in DMF (20:80).
`12. Kaiser test solutions (ninhydrin, pyridine, phenol) (see Note 1).
`13. Fmoc-amino-acids with protected side-chains (see Table 1).
`14. Trifluoroacetic acid (TFA).
`15. Triisopropylsilane (TIS).
`16.
`tert-butyl methyl ether (MTBE).
`
`(DMFJ, methanol
`
`methank
`[MeOH],dichloromedti
`
`3. Methods
`
`3.1. Principles of SPPS
`
`Steps, the useofas
`As peptide synthesis involves numerous repetitive steps, the
`support has obvious advantages. With such a system a large excess
`
`MPI EXHIBIT 1058 PAGE 8
`
`

`

`Modern Peptide Synthesis
`
`Table 1
`Proteinogenic Amino Acids
`
`5
`
`Side-chain
`H-
`CH3-
`(CH)),CH-
`(CH))CHCH,-
`CH,CH,(CH,)CH-
`HOOC-CH-
`H,NOC-CH,-
`HOOC-CH,CH,-
`H,NOC-CH,CH,-
`HN-CH,CH,CH,CH;-
`
`NH-CHCH;CH
`
`H,N
`
`CH,
`HO-CH,-
`CH-CH(OH)-
`
`-CHy
`
`HO CH
`ÇH-
`
`HS-CH;-
`CH,-S-CH,CHạ-
`
`One-letter code
`
`G A V L D N Q K R
`
`H
`
`S T
`
`Y
`
`W
`
`C M P
`
`H,N OH
`HR
`
`Amino acid
`Glycine
`Alanine
`Valine
`Leucine
`Isoleucine
`Aspartic acid
`Asparagine
`Glutamic acid
`Glutamine
`Lysine
`Arginine
`
`Histidine
`
`Serine
`Threonine
`Phenylalanine
`
`Tyrosine
`
`Tryptophan
`
`Cysteine
`Methionine
`Proline
`
`Three-letter code
`Gly
`Ala
`Val
`Leu
`Ile
`Asp
`Asn
`Glu
`Gln
`Lys
`Arg
`
`His
`
`Ser
`Thr
`Phe
`
`Tyr
`
`Trp
`
`Cys
`Met
`Pro
`
`Note: All the 20 DNA-encoded or proteinogenic a-amino acids are of L stereochemistry.
`
`high concentration can drive coupling reactions to completion. Excess reagents
`and side products can be separated from the growing and insoluble peptide sim-
`ply by filtration and washings, and all the synthesis steps can be performed in the
`Samevessel without any transfer of material.
`The principles of SPPS are illustrated in Fig. 2. The N-protected C-terminal
`amino acid residue is anchored via its carboxyl group to a hydroxyl (or chloro)
`
`MPI EXHIBIT 1058 PAGE 9
`
`

`

`Amblardeta.
`
`OH +
`
`Linker
`
`Loading of theresin
`
`N
`
`Linker
`
`P
`
`HN
`
`Removal of thetemporary
`Na protectinggroup
`
`Linker
`
`P
`
`permanent side-chain protecting group
`
`temporaryurethane Na protecting group
`
`solid support
`
`activating group
`
`NH or O
`
`6
`
`P T
`
`A X
`
`SPPS cycles
`
`Coupling of thenext
`activated amino acid
`
`Linker
`
`n cydes
`
`Linker
`
`Linker
`
`HN
`T
`
`FAn+2 H
`
`Rx
`Px
`
`Pn:2
`
`HN
`
`n
`
`Fig. 2. Principles of SPPS.
`
`MPI EXHIBIT 1058 PAGE 10
`
`

`

`Modern Peptide Synthesis
`
`7
`
`or amino resin to yield respectively an ester or amide linked peptide that will
`ultimately produce a C-terminal acid or a C-terminal amide peptide. After load-
`ing the first amino acid, the desired peptide sequence is assembled in a linear
`fashion from the C-terminus to the N-terminus (the C→ Nstrategy) by repeti-
`tive cycles of N deprotection and amino acid couplingreactions.
`Side-chain functional groups of amino acids must be masked with permanent
`protecting groups (P,) that are stable in the reaction conditions used during pep-
`tide elongation. The a-amino group is protected by a temporary protecting group
`(T) that is usually a urethane derivative. The temporary protecting group (T) can
`be easily removed under mild conditions that preserve peptide integrity and
`reduce the rate of epimerization, which can occur via 5(4H)-Oxazolone forma-
`tion of the activated amino acid during the coupling step (2,3) as indicated in
`Fig. 3. The protective role of urethanes against epimerization also explains the
`predominanceof the C→N strategy.
`After coupling, the excess of reactants is removed by filtration and washings.
`The temporary N-terminal protecting group is removed allowing the addition
`of the next N-urethane protected amino acid residue by activation of its a-car-
`boxylic acid. This process (deprotection/coupling)
`is repeated until the desired
`sequence is obtained.
`In a final step, the peptide is released from the resin and
`the side-chain protecting groups (P) concomitantly removed.
`
`3.2. Fmo/tBu SPPS
`
`In SPPS, two main strategies are used: the Boc/Bzl and the FmoctBu approaches
`for T/P, protecting groups. The former strategy is based on the graduated acid
`lability of the side-chain protecting groups. In this approach, the Boc group is
`removed by neat TFA or TFA in dichloromethane, and side-chain protecting
`groups and peptide-resin linkages are removed at the end of the synthesis by
`treatment with a strong acid such as anhydrous hydrofluoric acid (HF). While
`this method allows efficient syntheses of large peptides and small proteins, the
`use of highly toxic HF and the need for special polytetrafluoroethylene-lined
`apparatus limit the applicability of this approach to specialists only. Moreover,
`theuse of strongly acidic conditions can produce deleterious changes in the struc-
`tural integrity of peptides containing fragile sequences.
`The Fmoc/Bu method (4) is based on an orthogonal protecting group strat-
`egy. This approach uses the base-labile N-Fmoc group for protection of the a-
`amino function, acid-labile side-chain protecting groups and acid-labile linkers
`that constitute the C-terminal amino acid protecting group. This latter strategy has
`the advantage that
`temporary and permanent orthogonal protections are removed
`by different mechanisms, allowing the use of milder acidic conditions for final de-
`protection and cleavage of the peptide from the resin. For all thesereasons, Fmoc-
`basedSPPS is now the method of choice for the routine synthesis of peptides.
`
`MPI EXHIBIT 1058 PAGE 11
`
`

`

`8
`
`Amblard etal.
`
`RO
`
`H
`
`Base
`
`RO
`R,
`
`RO
`
`RO
`
`Base
`
`ROʻ
`
`RNH2
`
`RO,
`
`NHR,
`
`R1
`
`Fig. 3. Epimerization by oxazolone formation.
`
`3.3. Solid Supports
`
`The matrix polymer and the linker can characterize solid supports (5).Often
`the term "resin" is improperly used in place of the linker system, ignoringthe
`fact that the matrix polymer is as important
`in supported chemistry asthesolu-
`tion phase (6). As hundreds of different
`resins are commercially available,somt
`of them carrying the same linkers, special care should be taken to properlychoose
`the most suitable linker for the synthesis.
`
`3.3.1. Matrix Polymers
`
`commonly usedtor
`resins are most
`Cross-linked polystyrene (PS) -based
`(corresponding t01
`routine SPPS. Beads of 200 to 400 mesh size
`distribution
`diameter of about 50 um) and a loading of 0.5 to 0.8 mmol/g present goodchar
`acteristics for polymer swelling in solvents such as DMF and DCM,diffusto
`of reactants into the polymer matrix, and accessibility
`of
`linker sitesburncu
`into the bead. For larger peptides (more than 25 amino acids) or more diit
`sequences,a lower loading is required (0.1-0.2 mmol/g).
`vlene
`Cross-linked polyamide (PA)-based resins and composite PS-polyethy)e
`ohys
`glycol (PEG)-based resins are much more hydrophilic supports exhibitingp
`levels
`ical properties different
`from PS resins at microscopic and macroscopicle
`aler-
`(7). These supports, often with a lower
`loading capacity, may representa
`uences
`native to standard cross-linked PS resins for the synthesis of difficultseque
`and large peptides.
`
`MPI EXHIBIT 1058 PAGE 12
`
`

`

`Modern Peptide Synthesis
`
`Table 2
`Type of SPPS Reaction Vessels
`
`9
`
`length
`
`Vessel
`(cm)
`
`Vessel diameter
`(cm)
`
`Max. resin weight
`(g)
`
`Working volume
`(mL)
`
`5
`
`11
`15
`
`2
`2.6
`3.4
`
`0.5
`2
`
`10
`40
`90
`
`3.4. Resin Handling
`
`3.4.1. SPPS Reaction Vessels
`
`SPPS can be performed in classical glass reaction vessels that can be made by
`glassblowers or purchased from manufacturers (Fig. 1). Alternatively, syringes
`equipped with PTFE or glass frits may also be used.
`Reaction vessel size should be in relation to the amount of resin used, accord-
`ing to Table 2.
`
`3.4.2. Solvents
`
`As 99% of coupling sites are not at the surface but inside the resin beads,
`swelling of beads carrying the growing peptide chain is essential for the opti-
`mal permeation of activated N-protected amino acids within the polymer matrix,
`thus improving coupling yields. Before starting the solid phase synthesis, the
`resin has to be swollen in an adequate solvent such as DCM or DMF for 20 to
`30 min (Protocol 1). For cross-linked polystyrene beads used in SPPS, DCM
`presents optimal swelling properties. For coupling steps, polar aprotic solvents
`such as DMF or N-methylpyrrolidone
`(NMP) are preferred to improve solubil-
`ity of reactants. Alcohols and water are not adequate solvents for PS resins (see
`Note 2). Nevertheless, methanol or isopropanol can be used during the washing
`steps (Protocol 2) to shrink PS resin beads. This shrinking will efficiently remove
`reactants in excess. After such treatment, PS beads should be swollen in DCM
`or DME.
`
`3.4.3. Stirring and Mixing
`
`It is not necessary to agitate the reaction vessel vigorously, as diffusion phe-
`nomena dictate the kinetic reaction in SPPS. Moreover, most types of resin beads
`used for peptide synthesis are fragile, so magnetic stirring is not recommended.
`An old rotary evaporator rotor can be used for stirring during coupling and depro-
`tection steps or alternately any apparatus enabling smooth agitation by rocking
`
`MPI EXHIBIT 1058 PAGE 13
`
`

`

`10
`
`Amblard et al.
`
`or vortexing is appropriate. As beads usually stick to glass, the importantcon-
`dition is that all surfaces of the reactor must be in contact with thereaction
`mixture during stirTing.
`
`3.4.4. Washing
`
`to remove soluble side products and theexcessof
`Washing steps are essential
`reactants used during coupling and deprotection steps. Filling the reactorwith
`solvent contained in a wash bottle and emptying it under vacuum is anappro-
`priate and simple method.
`If necessary, stirring and mixing of the resininthe
`washing solvent can be performed with a PTFE stick.
`
`PROTOCOL 1. RESIN SWELLING
`
`1. Place the dry resin in the appropriate reaction vessel (see Subheading 3.4.1.).
`2. Fill the reactor with DCM until all
`resin beads are immersed. Resinsuspension
`can be gently mixed with a PTFE stick.
`3. Leave for 20 to 30 min.
`4. Remove DCM by filtration under vacuum.
`
`PROTOCOL 2. STANDARD WASHING PROCEDURES
`
`1. Fill the reaction vessel with DMF.
`filtration.
`2. Leave for 10 s and remove the solvent by
`3. Carefully wash the screw cap and the edge of the reactor with DMF.
`4. Repeat steps 1 and 2 twice with DME.
`5. Repeat steps 1 and 2 with MeOH.
`6. Repeat steps 1 and 2 with DCM.
`7. Repeat steps 1 and 2 with DMF.
`
`3.5. Linkers and Resins for Fmoc-Based SPPS
`
`d b
`
`loading) of theN-protected
`The first step of SPPS is the anchoring (or
`ond
`terminal amino acid residue to the solid support via an ester or an amidebo
`depending on the C-terminal
`functional group of target peptide(respectve
`nthe
`acid or amide). Most of the linkers are commercially available anchoredont
`different matrices (PS, PA, PEG-PS). The bead (ball) symbol used in thetolo
`ing paragraph is generic and does not refer to a particular matrix.
`
`3.5.1. Peptide Amides
`
`resins
`For the synthesis of C-terminal peptide amides, the commonly useuand
`are I to 3 (Fig. 4)
`(8-l0). These resins are compatible with Fmoc chemisty
`esidue,
`final TFA cleavage. For attachment of the first urethane N-protectedresu
`standard peptide coupling procedures (Protocol 5) can be used. Theseresl
`usually supplied Fmoc-protected and should be deprotected beforeinco
`
`MPI EXHIBIT 1058 PAGE 14
`
`

`

`Modern Peptide Synthesis
`
`11
`
`OMe NHạ
`
`OMe
`
`H,N
`Me0
`
`1: Rink am ide resin
`
`2: Pal resin
`
`3: Sieber resin
`
`HO
`
`Fig. 4. Resins for peptide amide synthesis.oOMe
`
`Me0
`
`HO
`
`4: X = H, Wang resin
`5: X = OMe, Sasrin resin
`
`6: HMPB resin
`
`7:X=H, Tritylchorideresin
`8: X =Cl 2- Chlorotrityl choride resin
`
`Fig. 5. Resins for peptide acid synthesis.
`
`tion of the first residue. With bulky C-terminal amino acids, a double coupling
`step can be necessary.
`
`3.5.2. Peptide Acids
`
`The anchoring of an amino acid to the solid support by esterification is often
`more difficult, and even hazardous,
`for some residues and can lead to epimer-
`ization, dipeptide formation, and low substitution. Thus, we recommend the
`purchase of resins preloaded with the first C-terminal N-protected amino acid;
`these are commercially available from various manufacturers.
`Commonly used resins in Fmoc/tBu strategy for the synthesis of C-terminal
`peptide acid are reported in Fig. 5 (11-14). Anchoring reactions must be per-
`formed in an anhydrous medium and amino acids containing water should be
`dried before use.
`
`3.5.3. Hydroxymethyl-Based Resins
`
`For hydroxymethyl-based resins 4 to 6, formation of the ester linkage is easier
`with unhindered resins such as Wang resin 4 compared with resins possessing
`withdrawing methoxy groups 5 and 6. The most commonly used esterification
`process is the symmetrical anhydride method (Protocol 3). Determination of
`the loading can be performed by Fmoc release measurement (see Note 3). In the
`case of difficult anchoring,
`the esterification step can be repeated with fresh
`reactants. Arginine derivatives can need three esterification steps to achieve
`
`MPI EXHIBIT 1058 PAGE 15
`
`

`

`12
`
`Amblard et al.
`
`corect loading. After anchoring, unreacted resin-bound hydroxyl groupsshould
`be capped by benzoic anhydride or acetic anhydride (see Protocol 3, step8).
`
`PROTOCOL 3. ATTACHMENT TO HYDROXYMETHYL-BASED RESIN
`
`1. Place the resin in the appropriate SPPS reactor.
`2. Swell
`the resin as described in Protocol 1.
`is placedinadry,
`3. The desired Fmoc amino acid (10 eq relative to resin substitution)
`round-bottom flask with a magnetic stirrer and dissolved in dry DCM at 0°C(3mU
`mmol). Some drops of dry DMF may be useful
`to achieve completedissolution.
`4. Add DIC (5 eq) and stir the mixture for 10 min at 0°C.
`If a precipitate isobserved,
`add DMF until dissolution and stir for 10 min longer.
`5. Add the solution to the hydroxyImethyl
`resin.
`6. Dissolve
`dimethylaminopyridine
`(DMAP)
`(0.1-1 eq) in DMF and add thesolu-
`tion to the reaction mixture.
`7. After 1 h stirring, wash the resin with DMF (three times) and finally withDCM.
`8. Dry the resin in vacuo for 18 h before
`performing Fmoc release measurementon
`a sample (see Note 3). When the loading is less than 70% repeat theesterification
`step.
`9. When the desired substitution is achieved, cap the remaining hydroxyl groupsby
`adding benzoic or acetic anhydride (5 eq) and pyridine (1 eq) in DMF totheresin
`(previously swelled) and stir for 30 min.
`10. Wash the resin (Protocol 2) and start classical elongation with N-protectedamino
`acid after Fmoc deprotection (Protocol 7).
`
`3.5.4. Trityl-Based Resins
`
`Trityl-based resins are highly acid-labile. The steric hindrance of thelinker
`prevents diketopiperazine formation and the resins are recommendedfor o
`and Gly C-terminal peptides. Extremely mild acidolysis conditionsenablete
`cleavage of protected peptide segments from the resin. These resins arecommer
`cially available as their chloride or alcohol precursors. The trityl chlorideresl
`is extremely moisture-sensitive, so reagents and glassware should becareruly
`dried before use to avoid hydrolysis into the alcohol
`form.
`It isnecessary
`tivate
`activate the trityl alcohol precursor and it is highly recommended toreacti
`ofthe
`the chloride just before use (see Note 4). After activation, attachmento
`pres
`first residue occurs by reaction with the Fmoc amino acid derivative inthe
`isfiee
`ence of a base. This reaction does not
`involve an activated species, soit1S
`from epimerization. Special precautions should be taken for Cys andHIs
`dues that are particularly sensitive to epimerization during activation (Tabe
`
`PROTOCOL 4. ATTACHMENT TO TRITYL-BASED RESIN
`
`1. Place 1 gof
`vessel.
`
`trityl-based resin (1.0-2.0 mmol chloride/g resin) in anSPPSI
`
`action
`
`MPI EXHIBIT 1058 PAGE 16
`
`

`

`Modern Peptide Synthesis
`
`13
`
`the resin as described in Protocol 1.
`2. Swell
`3. Add a solution of 3 eq of Fmoc amino acid and 7.5 eq of DIPEA in dry DCM
`(10 mL/g resin). When a lower substitution resin is desired, reduce the amount of
`amino acid.
`4. Stir the mixture for 30 to 60 min at room temperature.
`5. Wash the resin with DMF (two to three times).
`6 Add 10 mlL of a mixture of DCM/MeOH/DIPEA (80:15:5) to cap any remaining
`reactive chloride group.
`7. Mix for 15 min and filter.
`8. Wash the resin three times with DMF and DCM. After drying in vacuo, the substi-
`tution can be measured from Fmoc release (see Note 3).
`
`.6 Side-Chain Protecting Groups
`
`limit the description of side-chain protecting groups (5) to those that
`We will
`have been found most effective for the preparation of a large number of classi-
`cal peptide sequences in Fmoc SPPS and are commercially available from most
`of the protected amino acid providers. For routine synthesis, TFA-labile protect-
`ing groups are usually used. However,
`for selective modifications of a partic-
`ular residue on the solid support (e.g., side-chain cyclized peptides, biotinylated
`peptides), special orthogonal protecting groups are needed (15). Some of the
`most commonly used side-chain protecting groups are reported in Table 3.
`
`3.7. Coupling Reaction
`
`The most simple and rapid procedure for the stepwise introduction of N-pro-
`tected amino acids in SPPS is the in situ carboxylic function activation (Proto-
`col 5). A large excess of the activated amino acid is used (typically 2-10 times
`excess compared to the resin functionality, which is provided by the manufac-
`turer or empirically determined; see Note 3). This excess allows a high concen-
`tration of reactants (typically 60-200 mM) to ensure effective diffusion. The
`time required for a complete acylation reaction depends on the nature of the
`activated species, the peptide sequence that is already linked to the resin, and
`the concentration of reagents. This last parameter must be as high as possible
`and is in connection with the volume of the reaction vessel and the resin substi-
`tution (Table 2). The preferred coupling reagents for in situ activation are ben-
`zotriazol-1-yl-oxytripyrrolidino phosphonium hexafluorophosphate (PYBOP)
`(16) for phosphonium-based activation and O-(benzotriazol-1-yl)-1,1,3,3-
`tetramethyluronium tetrafluoroborate (TBTU) (17) or N-[1H-benzotriazol-1-yl)
`(dimeth-ylamino)methylene}-N-methyl-methanaminium hexafluorophosphate
`N-oxide (HBTU) (18) for aminium/uronium-based activation (Fig. 6). These
`coupling reagents convert N-protected amino acids into their corresponding OBt
`esters. A tertiary amine (generally disopropylethylamine)
`is required to produce
`
`MPI EXHIBIT 1058 PAGE 17
`
`

`

`14
`
`Amblardet l
`
`Table 3
`Side-Chain Protecting Groups in Fmoc-Based SPPS
`
`Protecting
`groups
`
`Pmc
`Pbf
`
`Removal
`condition
`
`95% TFA
`95% TFA
`
`Remarks and side reactions
`
`Presence of thiols may
`accelerate the cleavage.
`
`OtBu
`OAI|4
`
`95% TFA
`Pd(Ph;P)4/PhSiH,
`
`Aspartimide formation
`(see Subheading 12.2.).
`
`Amino acid
`and side-chain
`functionality
`
`Arg
`
`HN NH2
`
`Asp/Glu
`
`OH
`Asn/Gln
`
`NH2
`
`Cys
`nSH
`
`His
`
`Trt
`
`95% TFA
`
`Trt
`
`95% TFA
`
`Trt (NHr)
`Mta
`
`TFA
`1% TFA
`
`Protections avoiddehydration
`of the carboxamideside-chain
`during
`activation and helpto
`solubilize
`Fmoc-Asn-OH and
`Fmoc-GIn-OH.
`PyrGlu formation forN-terminal
`glutamine peptides
`(see Note 5).
`High level of epimerizationcan
`occur during activation
`(see Note 6).
`Participate in the folding of
`peptides and proteins by
`disulfide bridge formation
`(see Subheading 3.11.).
`
`Even with protection ofthe
`imidazole ring,problems
`of epimerization canoccr
`during activation.
`
`Trp can be usedunprotecleuo
`However, ifArg(Pme)
`Arg(Pbf)
`is
`esent inthecan
`Sequence, side-reaction
`Occur.
`
`Lys
`mNH2
`
`Ser/Thr/Tyr
`mOH
`
`Trp
`
`Boc
`Mtta
`Aloca
`tBu
`Trte
`
`Boc
`
`TFA
`1% TFA
`Pd(Ph,P)4JPHSİH,
`TFA
`1% TFA
`
`TFA
`
`"Protection used for on-resin derivatization.
`
`MPI EXHIBIT 1058 PAGE 18
`
`

`

`Modern Peptide Synthesis
`
`15
`
`(H,C),N O_N(CH)l2
`
`(H,0),N N(CH)2
`
`PF
`
`PYBOP
`
`BE
`
`O_
`TBTU
`
`HATU
`
`(HsC)N
`
`N(CH)2
`
`PE
`
`PPF,
`
`PYAOP
`HBTU
`Fig. 6. Phosphonium and uronium/aminium coupling reagents.
`
`Me
`Me-Nt
`
`MeN
`Me
`
`-HNa
`
`Fig. 7. N-terminal
`
`tetramethylguanidinated peptides.
`
`the carboxylate of N-protected amino acids which reacts with coupling reagents.
`More recently,
`N-[1H-benzotriazol-1-yl(dimethylamino)methylene]-N-methyl-
`methanaminium hexafluorophosphate (HATU)
`(19) and 7-azabenzotriazol-
`lyl-oxytris(pyrrolidino) hosphonium hexafluorophosphate (PYAOP) (20).
`that generate OAt esters, have been reported to be more efficient and to reduce
`epimerization.
`Uronium/aminium-based reagents (HBTU, HATU, TBTU) are thought to mech-
`anistically function in a similar way to their phosphonium analogs but unlike them,
`hey can irreversibly block the free N-terminal amino function of the peptide-
`Dy Tormingtetramethylguanidiniumderivatives(Fig. 7) (21). To avoid
`s side-reaction, it is recommended to generate the carboxylate of the amino
`acid before adding these coupling reagents at a slightly reduced equivalent com
`pared to the amino acid.
`
`MPI EXHIBIT 1058 PAGE 19
`
`

`

`16
`
`Amblard et al.
`
`PROTOCOL 5. STANDARDCOUPLING PROCEDURE WITH HBTU
`
`1. After Fmoc deprotection and washings (or properly conditioning andswellingwa
`the resin is dry), wash the resin once with DME.
`(usually 3 eq).
`2. Add the N-a Fmoc-protected
`amino acid as a powder
`3. Fill the SPPS reaction vessel (at least 2/3 of volume) with DMF and stir withaPIFE
`stick for 10-20 s.
`slight excess, 3.5-4 eq) and stir with aPIFE
`(usually
`4. Add DIPEA to the vessel
`stick until complete dissolution of the N-protected amino acid occurs.Otherteri:
`ary amines can be used such as N-methyl morpholine or triethylamine.
`5. Add HBTU (2.9 eq compared with 3 eq of amino acid), screw the cap, andstirfor
`30 min. For difficult sequences it is recommended to preactivate theN-protected
`amino acid before coupling to the free N"-amino function to limit theguanylaia
`side-reaction.
`6. Remove the coupling solution by filtration.
`7. Wash the resin properly according to Protocol 2.
`8. Perform qualitative monitoring of the coupling reaction using acolorimetrictes
`(see Note 1).
`
`test willreved
`In some cases coupling is not complete and a colorimetric
`the presence of free amino groups. On these occasions a double couplingwih
`fresh reagents must be performed. When
`acylation is incomplete after aseconl
`coupling, a capping procedure through acetic anhydride (Protocol 6)can
`performed to stop the elongation of these less-reactive amino groups.When
`colorimetric test still reveals the presence of free amine functions aftercapply
`aggregation can be suspected.
`
`PROTOCOL 6. CAPPING
`
`the resin in DCM.
`1. Swell
`2. Remove the DCM by filtration.
`3. Fill the SPPS reactor (at least 2/3 of volume) with a 50/50 DCM/aceticanhyu
`solution (in case of trityl
`linker, see Note 7) and mix for 3 min.
`4. Remove the capping solution by filtration and repeat step 3 for 7 min.
`5. Wash three times with DCM.
`etrice
`6. Check the disappearance of free amino groups by a convenient colorimeui
`(see Note 1) and repeat the operation if necessary.
`
`3.8. Difficult Sequences and Aggregation
`goW
`When N-deprotection reactions and amino acid coupling stepsdoo
`shoull
`completion or proceed in low yields, repeated or prolonged reactiontine
`overcomethese problems. Nevertheless, this is not always sufficient.Te
`of this failure is thought to be self-association of the peptide chainby nk
`bond formation leading to aggregation. This aggregation results in
`
`MPI EXHIBIT 1058 PAGE 20
`
`

`

`Modern Peptide Synthesis
`
`17
`
`Fmoc,
`
`OH
`
`Me0
`O-Fmoc
`Fig. 8. Fmoc-(FmocHmb) amino acid.
`
`solvation of the peptide-resin and inaccessibility of the reagents to the N-termi-
`nal amino group. This phenomenon is sequence-dependent with particular pro-
`pensity for sequences containing high proportions of hydrophobic residues and
`can start from the fifth coupled residue. Two general methods can be used to
`disrupt the formation of secondary structures. The first consists of changing the
`peptide environment by adding chaotropic salts such as potassium thiocyanate
`or lithium chloride at a 0.4 M concentration, detergent solvents at 1% (v/v), or
`solvents such as DMSO,
`trifluoroethanol, or hexafluoroisopropanol
`to the reac-
`tion medium (22-28). The second approach involves structural changes in the
`peptidebackbone itself by the introduction of protecting groups to selected amide
`bonds. This is usually achieved by using the 2-hydroxy-4-methoxybenzyl
`(Hmb)
`derivative of glycine. Other amino acids can also be incorporated as their N,O-
`bis(Fmoc)-N-(Hmb) derivatives approximately every 6-7 residues (Fig. 8) (29,
`30). The incorporation of (Hmb) amino acids can be performed through their
`corresponding pentafluorophenyl esters, which are commercially available, or
`by standard coupling methods. For optimal N-acylation of the terminal Hmb
`residue that can be slow,
`it is recommended to use powerful coupling methods
`suchas symmetrical anhydrides or preformed acid fluorides in DCM. The 0-
`Fmoc protection of the Hmb derivatives is cleaved under piperidine treatment
`andthe Hmb group in the final TFA cleavage. For Cys-, Ser-, and Thr-contain-
`ing peptides their pseudoproline derivatives, also known to disrupt aggregation,
`can also be used (31-33).
`
`3.9. Fmoc Deprotection
`
`Removal of the temporary Fmoc protecting group from the N-terminus of the
`peptidyl-resin is normally achieved by short treatment with 20% piperidine in
`DMF (Protocol 7). The reaction is generally complete within 10 min, but can
`be longer in some cases. For safe removal a 20-min deprotection time is recom-
`mended. The deprotection results in formation of a dibenzofulvene-piperidine
`adduct that strongly absorbs in the UV range (see Note 3). Fmoc removal can
`be monitored by UV spectroscopy. This is a common procedure with automatic
`
`MPI EXHIBIT 1058 PAGE 21
`
`

`

`18
`
`Amblard etal.
`
`synthesizers but not in standard manual SPPS. When incompletedeprotection
`is suspected, the use of 20% piperidine
`containing 1 to 5% DBU in DMF isteç.
`ommended. However, DBU can promote
`aspartimide
`formation,
`thus itsusg
`should be avoided in Asp- or

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