WO2006045603A1 - S-alkyl-sulphenyl protection groups in solid-phase synthesis - Google Patents

S-alkyl-sulphenyl protection groups in solid-phase synthesis Download PDF

Info

Publication number
WO2006045603A1
WO2006045603A1 PCT/EP2005/011476 EP2005011476W WO2006045603A1 WO 2006045603 A1 WO2006045603 A1 WO 2006045603A1 EP 2005011476 W EP2005011476 W EP 2005011476W WO 2006045603 A1 WO2006045603 A1 WO 2006045603A1
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
resin
group
alkyl
cysteine
Prior art date
Application number
PCT/EP2005/011476
Other languages
French (fr)
Inventor
Stéphane VARRAY
Oleg Werbitzky
Thomas Zeiter
Original Assignee
Lonza Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lonza Ag filed Critical Lonza Ag
Priority to EP05810158A priority Critical patent/EP1807442A1/en
Priority to AU2005298840A priority patent/AU2005298840B2/en
Priority to US11/666,171 priority patent/US20080108790A1/en
Priority to BRPI0518390-1A priority patent/BRPI0518390A2/en
Priority to JP2007538330A priority patent/JP2008529969A/en
Priority to CA002584906A priority patent/CA2584906A1/en
Priority to CN2005800365194A priority patent/CN101072791B/en
Publication of WO2006045603A1 publication Critical patent/WO2006045603A1/en
Priority to IL182751A priority patent/IL182751A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/655Somatostatins
    • C07K14/6555Somatostatins at least 1 amino acid in D-form
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • C07K1/061General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using protecting groups
    • C07K1/067General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using protecting groups for sulfur-containing functions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • C07K1/08General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using activating agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • C07K1/08General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using activating agents
    • C07K1/086General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using activating agents containing sulfur
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2/00Peptides of undefined number of amino acids; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids

Definitions

  • the present invention relates to a method of on-resin disulfide-bond formation in solid phase peptide synthesis (SPPS), and to respective peptide solid-phase conjugates.
  • SPPS solid phase peptide synthesis
  • protection groups can be employed for protection of cysteine residues, e.g. trityl, acetamidomethyl-, t-butyl, trimethylacetamidomethyl, 2,4,6-trimethoxybenzyl, methoxytrityl, t-butylsulphenyl.
  • trityl group is employed for simple protection during peptide synthesis.
  • cysteines that are subsequently subjected to cyclization by means of cystine formation
  • acetamidomethyl (acm)- protection group along with iodine oxidation has been most widely employed (Kamber et al., 1980, HeIv. Chim. Acta 63, 899-915; Rietman et al., 1994, Int. J. Peptide Protein Res. 44, 199-2O6).
  • iodine oxidizing susceptible side moities chain elsewhere, too.
  • Met may suffer from using iodine.
  • oxidation with iodine may set free HI, the acid then eventually promoting deprotection of side chains and/or, most importantly, cleavage from resin. Therefore the method must be applied as a late finishing step in synthesis only, after cleavage from resin, if used at all.
  • 91 to 114 glutathione in aequeous buffer, DMSO, potassium ferricyanide, Ellrnan's reagent , 5,5'- dithiobis(2-riitrobenzoic acid), iodine, thallium (III)trifiuoroacetate, alkyltrichlorosilane- sulphoxide, silver trifluoromethanesulphonate-DMSO mediated oxidation in strongly acidic medium.
  • all those methods give rise to undesireable, multiple side-products, require extended reaction times in the range of 10-20 hours for optimum yield and hence give ample opportunity to undesireable side-reactions.
  • US6476186 devises intramolecular disulfide bonding of an octapeptide in acetonitril/water (1:1) in the presence of trace amounts of charcoal.
  • the peptide was synthesized on 2- chlorotrityl resin and comprises apart from hydrophobic residues and the cysteines, a lysine and a threonine. Cysteines were protected with acid-labile trityl groups. Charcoal catalyzed cyclization took place after cleavage and deprotection in the aequeous solvent mixture.
  • the peptide according to the present invention may be any peptide comprising natural or non-natural amino acids such as e.g. homocysteines which homocysteines are preferably comprising 2-15 methylene groups and. one thiol group in their side chains , homoarginine, D-cyclohexyl-alanine, ⁇ -lysine, ⁇ -lysine, Penicillinamide (Pen) or ornithine (Om-) or D- analogues of the natural L-amino acids.
  • the peptide comprises only natural amino acids or the D-analogues or the homo- or nor-anlogues thereof.
  • peptide backbone or main chain, side chain and the prefixes 'nor-' 'homo-' are construed in the present context in accordance the IUPAC-IUB definitions (Joint IUPAC-IUB Corrirnission on Biochemical Nomenclature, 'Nomenclature and symbolism for amino acids and Peptides', Pure Appl. Chem., 56, 595-624 (1984).
  • 'homo-' and 'nor-' amount to just one extra or missing, respectively rnethylen bridging group in the side chain portion, preferably with the exception of homocysteines which may be defined preferably as said above.
  • su-ch further sulfhydryl-moiety comprising residues are protected by trialkylphosphine non- sensitive-, more preferably by tri-n-butylphosphine insensitive, protection groups, more preferably, such non-sensitive sulfhydxylprotection group is selected from the group comprising trityl-, tert. butyl-, acetamidomethyl-, alkylated acetamidomethyl-, alkylated trityl- protection groups.
  • side chain protection groups as commonly employed in the art (see e.g. Bodansky, M. , Principles of Peptide Synthesis, 2 nd ed. Springer Verlag Berlin/Heidelberg, 1993) may be used to protect susceptible side chains which could otherwise be modified in the coupling and deprotection cycles.
  • amino acids with susceptible side chains are Cys, Asp, GIu, Ser, Arg, Homo-Arg, Tyr, Thr, Lys, Orn, Pen, Trp, Asn and GIn.
  • a post solid-phase synthesis chemical modification of the peptide amide may be carried out to yield a desired side chain.
  • homoarginine can be prepared by guanidation of a lysine residue comprised in the peptide chain or an arginine can be prepared by guanidation of an ornithine residue comprised in the peptide chain. This may be a less viable option though in view of the additional reaction steps required.
  • coupling e.g. of Har requires extended coupling times and replenishing of coupling reagents.
  • Arg or Har preferably when being used as FMOC- Arg and FMOC- Har respectively, without the use of side chain protecting groups.
  • This may be achieved by ensuring that post-coupling of the individual Arg or Har residue, the guanidino moiety is quantitatively protonated prior to any further coupling reactions and forms stable ion pair with the proton donor in organic solvent.
  • This is preferably achieved by treating the resin bound peptide amide with an excess of the acidic coupling auxilliary BtOH or the like as described in more detail below in the experimental section.
  • Another example of scavenging the charge of the guanidinh ⁇ m group is to use tetraphenyl borate salts of Fmoc- protected HAR for synthesis as set forth, in US 4,954,616.
  • the solid phase support or resin may be any support known in the art that is suitable for use in solid-phase synthesis.
  • This definition of solid phase comprises that the peptide is bonded or linked via a functional linker or handle group to the solid phase or resin.
  • the solid support is based on a polystyrene or polydimethylacrylamide polymer, as is customary in the art.
  • the peptide may be bonded via a suitable amino acid side chain, including e.g. the thiol moiety of a further cysteine residue of the peptide intended not to participate in the cyclization reaction, or may be bonded via the C-terminal ⁇ -carboxy group to a resin by means of e.g.
  • an ether, thioetXier, ester, thioester or amide bond examples are solid supports comprising handle groups such as e.g. trityl, 2-chloro-trityl-, 4-methoxytrityl-, 'Rink amide' 4-(2',4'-dimethoxybenzyl- aminomethyl)-phenoxy-, Sieber resin (9-amino-6-phenylmethoxy-xanthen-), A- hydroxymethylphenoxyacteyl-, 4-bLydroxymethylbenzoic acid (the latter requiring attachement of the first amino acid by means of p-dimethylaminopyridine-catalysed esterification protocol than can result in racemisation of susceptible amino acids, e.g.
  • thioester linkages for bonding of the peptide moiety to the solid- phase are specifically disclaimed since subject to transthioesterification side reaction under at least slightly basic pH.
  • Thioester linkages are vulnerable to treatment with S-tert.butyl-sulphenyl protection group removing agents, in particular those of the thiol reducing type such as ⁇ -mercapto-ethanol in near- stochiometric amounts or beyond.
  • the S-tert.butyl-sulphenyl protection group removing agent is a phosphine, more preferably a tris-(Cl-C8) alkyl-phosphine wherin the alkyl may be, independently, further substituted with halogeno or (Cl-4)alkoxy or (Cl-C4)ester. More preferably, the removing agent is a tris-(C2-C5)alkyl-phosphine wherein the alkyl may be further substituted,independently, with (Cl-C2)alkoxy.
  • S-S-bond-comprising resin handles such as the HPDI bifunctional hydroxy and disulfide handle described in Brugidou, J. et al., Peptide Research (1994) 7:40-7 and Mery, J. et al, Int. J. Peptide and Protein Research (1993), 42: 44-52) are of course excluded from the scope of the present invention since not allowing of on-resin cyclization.
  • On-resin cyclization according to the present invention allows of avoiding the problems arising from intermolecular side reaction and the dilution techniques or catalyst-surface absorption techniques usually employed for this reason.
  • Rink amide, Sieber resin Tetrahedron Lett. 1987, 28, 2107-2110 or similiar 9-amino- xanthenyl-type resins
  • PAL resins Albericio et al., 1987, Int. J. Pept. Protein Research 30, 206-216
  • the specially substituted trityl-amine derivatives according to Meisenbach et al., 1997, Chem. Letters , p. 1265 f.
  • the peptide is anchored to the solid phase by either an amide or ester bond via the C-terminus.
  • the solid phase is an acid-sensitive or acid-labile solid phase, even more preferably, it is an amide generating acid-labile solid-phase.
  • acid- labile solid phases require at least 0.1% trifluoro acetic acid (TFA), more preferably at least 0.5% TFA in a polar aprotic solvent for cleavage from resin.
  • the solid- phase is an acid-sensitive solid ptiase that is cleaved under weakly acidic conditions, that is 0.1 to 10% TFA in said solvent are sufficient to effect at least 90% cleavage efficiency upon incubation at room temperature up to 5 hours.
  • Such highly acid-labile solid phase are e.g. 2-chlorotrityl resins, 4,4'-dirnethoxytrityl resin, the related, trityl-based phenylalcolhol resin such as e.g.
  • NovasynTM TGT derived from an conventional aminomethyl resin by acylation with Bayer's 4-carboxytrityl linker or a 4-methoxyphenyl, 4,4'-dimethoxyphenyl or 4-methyl-derivative of said linlcer, further Sieber resin, Rink amide resin or 4-(4- hydroxymethyl-3-methoxyphenoxy)-butyric acid (HMPB) resin, (4-methoxybenzhydryl-) or (4-methylbenzhydryl)-phenyl resins, the former said Sieber and Rink resin specifically giving rise to C-terminally amidated peptide upon acido lysis.
  • HMPB 4-(4- hydroxymethyl-3-methoxyphenoxy)-butyric acid
  • Such acid-labile solid phases are particurlarly vulnerable to on-resin deprotection chemistries for side-chain protection groups and hence particular attention must be paid in these cases.
  • the linking bond In case of side chain anchoring via C-terminal cysteine residue to the handle group of a solid support, the linking bond must be a thioether or thioester bond.
  • Further suitable residues for side-chain anchoring are carboxy groups of acidic side chains, hydroxy groups and in particular the ⁇ -amino group of lysine. It goes without saying that in case of side chain anchoring, that the C-terminal free carboxygroup is generally to " be protected by esterification or amidation prior to carrying out the first coupling reaction, e.g. by using FMOC-Lys-carboxamid for linking reaction of the side chain amino function to the solid phase.
  • one S-alkyl-sulphenyl-protected cysteine preferably one S- tert.butyl-sulphenyl protected cysteine is the C-terminal residue of the peptide and is bonded via the carboxy-terminus by means of an ester or amide bond to the solid phase, with the proviso, that said linking bond is not a benzylester moiety but preferably is an acid-labile resin that is cleaved under weakly acidic reaction conditions as defined above.
  • a C-terminal cysteine is particularly prone to subject to racemisation in acidic conditions, e.g. upon cleavage and/or deprotection under strongly acidic condition.
  • heterogenous catalysts for air-borne, oxidative cyclization are e.g. charcoal, which is incompatible ⁇ vith use on a solid-phase. It may not be efficiently removed. Preferably, it relates to the absence of a catalytically effective or substantial amount of such heterogenous catalyst. Not using inappropriate catalyst when not required for the purposes of the present invention is a self-evident measure to the skilled artisan, though.
  • Coupling reagents for peptide synthesis are well-known in the art (see Bodanslcy, M. , Principles of Peptide Synthesis, 2 nd ed. Springer Verlag Berlin/Heidelberg, 1993 ; esp. cf. discussion of role of coupling additivesauxilriari.es therein).
  • Coupling reagents may be mixed anhydrides (e.g. T3P: propane phosphonic acid anhydride) or other acylating agents such as activated esters or acid halogenides (e.g. ICBF, isobutyl-chloroformiate) , or they may be carbodiimides (e.g.
  • the coupling reagent is selected from the group consisting of uronium salts and phosphonium salts of the benzotriazol capable of activating a free carboxylic acid function along with that the reaction is carried out in the presence of a base.
  • coupling salts are e.g. HBTU (0-lH-benzotriazole-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), BOP (benzotriazole- 1 -yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate) , PyB OP (B enzotriazol- 1 -yl-oxy-tripyrrolidinopho sphonium hexafluorophosphate), PyAOP, HCTU (O-ClH- ⁇ -chloro-benzotriazole-l-yrj-U ,3, 3- tetramethyluronium hexafluorophosphate), TCTU (O-lH-6-chlorobenzotriazole-l-yl)- 1,1,3,3-tetramethyluronium hexafluorophosphate),
  • a further or second weak base reagent is needed for carrying out the coupling step.
  • base whose conjugated acid has a pKa value of from pKa 7.5 to 15, more preferably of from pKa 7.5 to 10, with the exclusion of an ⁇ -amino function of a peptide or amino acid or amino acid derivative, and which base preferably is a tertiary, sterically hindered amine.
  • H ⁇ nig-base N,N- diisopropylethylamine
  • N,N'-dialkylaniline 2,4,6-trialkylpyridine
  • N-alkyl-r ⁇ orpholine with the alkyl being straight or branched C1-C4 alkyl, more preferably it is N- methylmorphorine or collidine (2,4,6-trimethylpyridine), most preferably it is collldine.
  • coupling additives in particular of coupling additives of the benzotriazol type, is also known (see Bodansky, supra). Their use is particularly preferred when using the highly activating, afore said uronium or phosphonium salt coupling reagents.
  • the coupling reagent additive is a nucleophilic hydroxy compound capable of forming activated esters, more preferably having an acidic, nucleophilic N- hydroxy function wherein N is imide or is N-acyl or N-aryl substituted triazeno, most preferably the coupling additive is a N-hydroxy-benzotriazol derivative (or 1 -hydroxy- benzotriazol derivative) or is an N-hydroxy-benzotriazine derivative.
  • N-hydroxy compounds have been described in large and wide in WO 94/07910 and EP -410 182 and whose respective disclosure is incorporated by reference hereto. Examples are e.g. Kf-hydroxy-succinimide, N-hydroxy-3,4-dihydro-4-oxo-l,2,3- benzotriazine (HOOBt), l-hydroxy-7-azabenzotriazole (HOAt) and NT-hydroxy- benzotriazole (HOBt).
  • N-hydroxy-benzotriazine derivatives are particularly preferred, in a most preferred embodiment, the coupling reagent additive is hydroxy-3,4-dihyclro-4-oxo- 1,2,3-benzotriazine.
  • Ammonium salt compounds of coupling additives are known and their use in coupling chemistry has been described, for instance in US4806641.
  • the uronium or phosphonium salt coupling reagent is an uronium salt reagent and preferably is HCTU, TCTU or HBTU and even more preferably is used in the reaction in combination with N-hydroxy-3,4-dihydro-4-oxo- 1,2,3-benzotriazine or a salt thereof.
  • This embodiment is mainly preferred for use in chain elongation step of peptide synthesis after removal of the base-labile N ⁇ -protection group, but may as well be used for lactamization reaction during side-chain cyclization.
  • HCTU and TCTU are defined as to be encompassed by the term 'uronium salt reagent' despite that these compounds and possible analogues have been shown to comprise an isonitroso moiety rather than an uronium moiety by means of crystal structure analysis (O. Marder, Y. Shvo, and F.
  • the coupling reagent is a phosphonium salt of the benzotriazol such as e.g. BOP, PyBOP or PyAOP.
  • Deprotection of the base labile Na may be carried out as routinely done in the art, e.g. with 20% piperidine in N-methyl morpholine when using standard Fmoc chemistry.
  • Fmoc or Boc protection chemistry for the N-terminus is routinely applied in solid phase synthesis but further optional Na protection chemistries are known in the art and can be applied where not interfering with the present invention, that is to devise disullide-borne peptide cyclization of the resin-conjugated peptide.
  • S-alkyl-sulphenyl protecing groups protecting thiol groups of cysteine or homocysteine residues are removed according to the present invention by a reagent that typically is capable of removing, preferably substantially removing, the S-tert.butyl-sulphenyl-protection group from such residue.
  • a reagent typically is capable of removing, preferably substantially removing, the S-tert.butyl-sulphenyl-protection group from such residue.
  • Removal of S- tert.butyl-sulphenyl protection groups from e.g. cysteine accomplished by means of reaction with tertiary phosphines has been described, for instance by using tributylphosphine (Atherton et al., 1985, J. Chem. Soc, Perkin I.
  • tert- butylsulphenyl group is also cleaved in an orthogonal fashion by means of thiol reagents such as e.g. ⁇ -mercapto-ethanol or dithio-threitol (DTT) as an option to using tertiary phosphines (Huang et al.,1997 Int. J. Pept. Protein Res. 48, 290; Rietmann et al., 1985, Reel. Trav. Chim. Pays-Bas, 1141).
  • thiol reagents such as e.g. ⁇ -mercapto-ethanol or dithio-threitol (DTT)
  • the tertiary phosphine is triphenylphospine or is an (C1-C4) alkylated or (Cl-C4)alkoxylated triphenylphosphine, such as e.g. tri-(p- methoxyphenyl)-phosphine or even more preferably is a trialkylphosphine wherein the alkyl may be the same or different , and wherein each alkyl is a Cl to C7 alkyl, preferably Cl to C4 alkyl, and maybe branched or linear alkyl.
  • the alkyl is linear.
  • alkyl may be optionally further substituted with halogeno, (Cl-C4)alkoxy such as e.g. and preferably methoxy or ethoxy, or may be further substituted where amenable with the solvent system, carboxy or is, preferably, unsubstituted.
  • halogeno, (Cl-C4)alkoxy such as e.g. and preferably methoxy or ethoxy, or may be further substituted where amenable with the solvent system, carboxy or is, preferably, unsubstituted.
  • disulfide cleavage by means of phosphines may also be used with the acid-labile resins cleavable in weakly acidic reaction conditions such as Sieber or 2-chloro-trityl (CTC) resin, for instance. It is also often overlooked that thiol reagens reduce and hence cleave disulfides " by forming disulfide products themselves.
  • such thiol reagent is selected from the group consisting of erythro-2,3-dihydroxy-l,4-butanedifhiol (or named meso-1,4- Dithioerythritol or DTE for short ), DL ⁇ threo-2,3-dihydroxy-l,4-butanedithiol (or named rac-l,4-Dithiothreitol or DTT for short), L-threo-2,3-dihydroxy-l,4-butaneditliiol, D- threo-2,3-dihydroxy-l,4-butanedithiol and mixtures thereof.
  • Mixtures may comprise DTE and DTT, either in its racemic form or as an optically active preparation of DTT. More preferably, the thiol reagent is DTT which means D-,L- or any racemic or non-racemic mixture thereof. - DTT and DTE are also known as Cleland's reagent and Cleland's other reagent, respectively (Cleland, W. , Biochemistry 3,480-482,1964).
  • Cyclization is carried out according to the present invention in the presence of a first weak base in a polar, aprotic organic solvent in the presence of air and/or oxygen but notably in the absence of a heterogenous, rate-accelerating catalyst. Still then, and without precedent, the cyclization step, due to the method of the present invention, is remarkably efficient and requires only about 0.5 to 2 hours reaction time, allowing of literally quantitative, complete conversion of educt to the desired product under very mild reaction conditions (ambient temperature typically, expedient temperature range being 1O 0 C to 8O 0 C though reflux temperature of solvent must be taken into account of course). Conversion is complete.
  • Suitable polar, aprotic solvents are e.g. acetonitril, dimethylformamide, dichloxomethane, N-methyl-pyrrolidone, tetrahydrofurane. In contrast to water, such solvent us ⁇ ally may not physically dissolve relevant amounts of oxygen to supply the oxidative formation of disulfide bonds as has been described for aequeous catalyst systems before.
  • Air/oxgen maybe supplied by thorough stirring, vortexing, special design of propellers used for stirring, gas sparging into the liquid.
  • the gas may be air or pure oxgen or air enriched with oxgen which is vented or sparged into the reaction liquid.
  • large or surface areas of the bottom and/or walls of the reactor vessel are puntctured as to allow of sparging gas into the liquid, under thorough stirring.
  • the reaction vessel comprises a fritted bottom or a fritted section of least 50% surface area of the total surface of the bottom, as to allow of simultaneous stirring and venting by upsurging, bubbling air vented into the reaction liquid through that bottom.
  • the first weak base reagent is a weak base whose conjugated acid has a pKa v ⁇ alue of from pKa 7.5 to 15, more preferably of from pKa 8 to 10, preferably, it is a tertiary;, sterically hindered amine.
  • H ⁇ nig-Base N 5 N " - diisopropylethylar ⁇ ine
  • N,N-dialkyl-aniline 2,4,6-trialkylpyrididine
  • N-allcyl- morpholiiie with the alkly being straight or branched C1-C4 alkyl such as methyl, ethyl, n- propyl, i-propyl, n-butyl, most preferably it is N-methylmorpholine, collidine (2,4,6- trimethylpyridine) or H ⁇ nig-Base.
  • the prior removal of the disulfide-bonded protection group according to the present invention notably the removal of the S-tert.butyl-sulphenyl group is effected in the presence of a first -weak base reagent, for avoiding any risk of leakage from ttaie resin by minor acidolysis, tliat is at a pH of from 7.5 to 12, more preferably of from 8 " to 11.
  • polar aprotic solvents such as THF or acteonitril that are freely miscible with water
  • basic salts such as e.g. sodium acetate in aequeous solution may be used for that purpose.
  • This embodiment is particularly preferred when using tertiary phosphines for said, disulfide group cleavage or removal step.
  • a suitable oxygen supply concomittant with such disulfide protection group removal, it may be possible in another embodiment of the present invention, e.g. when using polar, aprotic organic solvent along with oxygen supply in the presence of a tertiary amine and when using tertiary pl ⁇ osphine for deprotection that is inert to oxygen, to carry out both disulfide deprotection and cyclization not only in a one-pot reaction but even as a single reaction step.
  • the on-resin operation mode of the invention allows of quick and efficient intra-molecular cyclization only, giving no chance of dimerization at all.
  • the peptide is the peptide of formula I or II.
  • protection group is to be construed as being protection group for a gi ⁇ ven side chain functionality or specific side chain which protection group is compliant with being used in standard tert-bixtyloxycarbonyl (Boc) or 9-fluorenylmethoxycarbonyl (Fmoc) solid phase peptide synthesis.
  • Boc tert-bixtyloxycarbonyl
  • Fmoc 9-fluorenylmethoxycarbonyl
  • a side chain group Rl (o) for instance it not to be construed in the way as to refer to a single type of optionally protected amino acid side chain; each residue Rl(I), Rl(2)...may be unique or may be the same as at least one other residue.
  • the peptide of formulas I and II may also comprise well-known peptide backbone modifications that are commonly employed in peptide synthesis: cyclic amino acids such as D- or L-Pro, intermittent non- peptide moieties linking two peptidyl segments and being e.g. hairpin or ⁇ -turn mimetics or in particular backbone-modified dipeptidyl segments used in synthesis e.g. for introducing amide protected Asp-Gly(Hmb) segments avoiding aspartimide formation (Packman et al. 1995, Tetrahedron Lett.
  • the two cysteines that are going to be disulfide-connected in cyclization are spaced apart by at least two amino acid residues (or the like).
  • a spacing of i+3 is typical of an ⁇ -lielical peptide conformation and allows of optimal,spacial juxtaposition for disulfide bonding. In this way, cyclization is facilitated.
  • the constraint exercised by the backbone in view of the possible, more stable conformations is rendering cyclization more difficult.
  • a peptide on a solid phase not by permanent, covalent attachment of the peptidyl moiety to a solid-phase but by non- covalent, reversible attachement to the solid-phase by means of a stable metal chelate complex (pressrelease October/November 2004 made by Lonza AG, Basel, Switzerland jointly with AplaGen GmbH, Baesweiler, Germany, October 20O4), similiar to the hexa- His tag technology employed in protein purification since long.
  • a further object of the present invention are the respective, solid-phase borne peptides or solid-phase-peptide conjugates, respectively.
  • the relevant definitions given above and belo ⁇ V apply likewise to such object, alone or in combination.
  • said further object of the present invention is a peptide of formula I or II,
  • Coupling time for loading was twice the standard coupling time, namely 60 min. in total. Couplings were conducted with 2 eq. each of respective amino acid in the presence of leq. each of 6- chloro-HOBt, TCTU, H ⁇ nig-Base (Disopropylethylamiae), in dichloromethane. Washes were with N-methyl-pyrrolidone (NMP).
  • NMP N-methyl-pyrrolidone
  • FMOC deprotection was done by 3 cycles of 15 min. 10*% piperidine in N-methyl- pyrrolidone; efficiency of cleavage and completion of synthesis was analysed by Ninhydrin reaction and reverse phase HPLC, respectively.
  • step 1.2 The resin product of step 1.2 was suspended and washed three times in tetrahydrofurane (THF). The reaction was carried out for 1 h at room temperature with 50eq. tributylphosphine made up as 19%(v/v) PBu 3 /77%(v/v) THF /4%(v/v) saturated aequeous solution of sodium acetate; precipitating salt was filtered off prior to use . Reaction proceeded uniformly to give one dominant product peak. The yield was determined by reverse phase HPLC and was found to amount to 98.9%correct product.
  • THF tetrahydrofurane
  • the conversion to product was 100%, as determined independently by both reverse phase HPLC and LC-MS.
  • RP-HPLC was carried out on a Hypersil-KeystoneTM Betabasic (Thermo Electron Corp., Waltham Mass./U.S.A.) Cl 8 150x4.6 mm column, with an injection volume of 15 ⁇ l and detection at 262nm at a column temperature of 35 0 C. Gradient run is
  • Reaction takes place at 15 0 C for 2 h on an slowly rotating orbital shaking device. Reaction is terminated and product is precipitated, after filtering off the resin, by dropwise addition of tert.butyric acid methyl ester. The product is a uniform peak; no major sideproduct can be detected.- the above conditons of global deprotection have been tested on a control and found not to affect preformed disulfide bridges in peptides.
  • DTT is either rac- or L-DTT, obtainable from Biosynth AG/Switzerland.
  • the resin product of step 1.2 was suspended and washed three times in dimethylformamide (DMF). 50 eq. of DTT were used, made up as DMF/DTT (1 :1) and the reaction time was extended to 3-5 hours at room temperature. Subsequently, the peptide-resin was treated exactly as described in section 1.4-1.6 above. Yields obtained perfectly matched that of 1.6, with similar purity.
  • Vapreotide a Somatostatin peptidagonist
  • a Somatostatin peptidagonist is synthesized essentially as described above in section 1.1. Further processing is carried out essentially as described in sections 1.2-1.6, providing the deprotected Vapreotide-carboxamide in excellent yield and purity. Optionally, deprotection according to section 2. is carried out, likewise with very good result. 4. Cvclisation of BOC-LVS(BOC)-CvS(S-IBu)-ASn-ThT(TIt)-AIa-ThXfTn)-CvSfS-IBu)-AIa-
  • Synthesis is essentially carried out as described in section 4. above, except that the last Lys residue is added after cyclization reaction in an additional coupling cycle, that synthesis is carried out on a Rink amide resin and that prior to global deprotection, cleavage is carried out under weakly or mildly acidic condition: Cleavage from resin is achieved with 3 cycles of 15 min. each at 15 0 C, 2% (w/w) TFA, 1% (w/w) triethylsilane (TES) in dichloromethane. The reaction is stirred by nitrogen bubbling. After each cycle, cleavage reaction is directly quenched by pouring the whole reaction broth into dilute pyridin
  • Synthesis and cyclization is essentially carried out as described in sections 1.1-1.5 above,except that 2-chlorotrityl-polystyrene resin (CBL Patras, Greece) is used as a solid phase and that the DTT method according to section 2 is used instead of 1.3/phosphine method. Further, cleavage under mildly acid condition without side chain deprotection is used, essentially as described in section 5. Good yields are obtained.
  • Fragment synthesis serves as an optional route to Pramlintide synthesis: the cylized, bridging-cystine comprising but still protected peptide is then subjected to conventional fragment coupling technique with a C-terminal, residual fragment of Pramlintide using standard peptide coupling chemistry with TCTU wherein the C-terminal, protected fragment is harbored either on solid-phase or, preferably, in liquid phase, too.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Endocrinology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

A novel method for on-resin formation of disulfide-borne cyclization of peptides is devised.

Description

S-alkyl-suIphenyl protection groups in solid-phase synthesis
The present invention relates to a method of on-resin disulfide-bond formation in solid phase peptide synthesis (SPPS), and to respective peptide solid-phase conjugates.
A large variety of protection groups can be employed for protection of cysteine residues, e.g. trityl, acetamidomethyl-, t-butyl, trimethylacetamidomethyl, 2,4,6-trimethoxybenzyl, methoxytrityl, t-butylsulphenyl.
Most commonly, the trityl group is employed for simple protection during peptide synthesis. For protection of cysteines that are subsequently subjected to cyclization by means of cystine formation, acetamidomethyl (acm)- protection group along with iodine oxidation has been most widely employed (Kamber et al., 1980, HeIv. Chim. Acta 63, 899-915; Rietman et al., 1994, Int. J. Peptide Protein Res. 44, 199-2O6). As a disadvantage, the spectrum of side-product impurities is substantially enhanced by using iodine, oxidizing susceptible side moities chain elsewhere, too. E.g. Tyr, Met may suffer from using iodine. More importantly, oxidation with iodine may set free HI, the acid then eventually promoting deprotection of side chains and/or, most importantly, cleavage from resin. Therefore the method must be applied as a late finishing step in synthesis only, after cleavage from resin, if used at all.
The prior art knows a multitude of oxidating agents, beside iodine, which are added for allowing of cystine formation (examples derived from Albericio et al., in: Chan and White, eds., 'FMOC Solid-phase Peptide Synthesis', Oxford university Press 2000, p. 91 to 114: glutathione in aequeous buffer, DMSO, potassium ferricyanide, Ellrnan's reagent , 5,5'- dithiobis(2-riitrobenzoic acid), iodine, thallium (III)trifiuoroacetate, alkyltrichlorosilane- sulphoxide, silver trifluoromethanesulphonate-DMSO mediated oxidation in strongly acidic medium. Usually, all those methods give rise to undesireable, multiple side-products, require extended reaction times in the range of 10-20 hours for optimum yield and hence give ample opportunity to undesireable side-reactions. Volkmer-Engert et al. (Surface- assisted catalysis of intramolecular disulfide bond formation in peptides , J. Peptide Res. 51, 1998, 365-369) describe charcoal-catalyzed oxidative formation of disulfide bonds in water by using oxygen dissolved in the solvent, i.e. water. Careful controls showed that the pool of oxygen physically dissolved in the aequeous medium was necessary and sufficient to load the charcoal with oxygen for oxidation. Use of charcoal, as compared to traditional air-sparging in the absence of catalyst, accelerated the reaction rate dramatically.
The use of charcoal inevitably requires to carry out such reaction in homogenous solution but not on-resin; subsequent reaction steps of deprotection would not tolerate the continued presence of charcoal which is impossible to remove from the peptide-resin solid phase though. Cyclization accordingly takes place after cleavage from the resin, that is in solution. Cleavage from the solid support and global deprotection prior to cyclization is mandatory in this scheme. As a further disadvantage, Atherton et al. (1985, J. Chem. Perkin Trans. I. , 2065) reported that the use of the popular both scavenger and acido lysis promoter thioanisol in acidic deprotection also resulted in partial, premature deprotection of acm, tert-butyl and tert-butylsulphenyl protected cysteines.
US6476186 devises intramolecular disulfide bonding of an octapeptide in acetonitril/water (1:1) in the presence of trace amounts of charcoal. The peptide was synthesized on 2- chlorotrityl resin and comprises apart from hydrophobic residues and the cysteines, a lysine and a threonine. Cysteines were protected with acid-labile trityl groups. Charcoal catalyzed cyclization took place after cleavage and deprotection in the aequeous solvent mixture.
It is an object of the present invention to devise a more simple and straightforward, other or improved method for synthesizing disulfide-bonded cyclic peptides by means of solid phase synthesis. This object is solved, according to the present invention, by a method of peptide synthesis comprising the steps of
a. synthesizing a peptide linked to a solid phase which peptide comprises at least two residues of a cysteine or a homo-cysteine, which cysteines are protected in their side chain each by a S-alkyl-sulphenyl protection group, wherein the alkyl may be further substituted with aryl, aryloxy, alkoxy, halogenated variants thereof or halogeno, and wtterein the two protection groups may be the same or different, preferably they are protected in their side chain each by a S-tert.butyl- sulphenyl group, and
b. further reacting the peptide Λvith a S-tert.Butyl-sulphenyl-protection grovxp removing reagent, preferably reacting the peptide with a tertiary phosphiαie, and
c. cyclizing the peptide by means of disulfide bond formation in the presence of air and/or oxygen but, preferably, in the absence of a heterogenous catal^yst.
The peptide according to the present invention may be any peptide comprising natural or non-natural amino acids such as e.g. homocysteines which homocysteines are preferably comprising 2-15 methylene groups and. one thiol group in their side chains , homoarginine, D-cyclohexyl-alanine, ε-lysine, γ-lysine, Penicillinamide (Pen) or ornithine (Om-) or D- analogues of the natural L-amino acids. Preferably, the peptide comprises only natural amino acids or the D-analogues or the homo- or nor-anlogues thereof. - The terms peptide backbone or main chain, side chain and the prefixes 'nor-' 'homo-' are construed in the present context in accordance the IUPAC-IUB definitions (Joint IUPAC-IUB Corrirnission on Biochemical Nomenclature, 'Nomenclature and symbolism for amino acids and Peptides', Pure Appl. Chem., 56, 595-624 (1984). In its more narrow and preferred meaning, 'homo-' and 'nor-' amount to just one extra or missing, respectively rnethylen bridging group in the side chain portion, preferably with the exception of homocysteines which may be defined preferably as said above.
Particular attention must be paid to further side-chain protection of the amino acids forming the peptidic sequence, in particular when referring to further cysteine, horrxo- or nor-cysteine residues comprised in the peptide sequence that are intented to remain protected during rather than to participate in the cyclization reaction. Preferably, su-ch further sulfhydryl-moiety comprising residues are protected by trialkylphosphine non- sensitive-, more preferably by tri-n-butylphosphine insensitive, protection groups, more preferably, such non-sensitive sulfhydxylprotection group is selected from the group comprising trityl-, tert. butyl-, acetamidomethyl-, alkylated acetamidomethyl-, alkylated trityl- protection groups.
On the more general level, side chain protection groups as commonly employed in the art (see e.g. Bodansky, M. , Principles of Peptide Synthesis, 2nd ed. Springer Verlag Berlin/Heidelberg, 1993) may be used to protect susceptible side chains which could otherwise be modified in the coupling and deprotection cycles. Examples of amino acids with susceptible side chains are Cys, Asp, GIu, Ser, Arg, Homo-Arg, Tyr, Thr, Lys, Orn, Pen, Trp, Asn and GIn. Alternatively, a post solid-phase synthesis chemical modification of the peptide amide may be carried out to yield a desired side chain. For instance, as set forth amply in different references (EP-301 850; Yajima et al, 1978, J. Chem. Cos. Chem. Commun., p.482; Nishimura et al., 1976, Chem. Pharm. Bull. 24:1568) homoarginine (Har) can be prepared by guanidation of a lysine residue comprised in the peptide chain or an arginine can be prepared by guanidation of an ornithine residue comprised in the peptide chain. This may be a less viable option though in view of the additional reaction steps required. Notably, coupling e.g. of Har requires extended coupling times and replenishing of coupling reagents. According to the present invention, it is one preferred embodiment to couple Arg or Har, preferably when being used as FMOC- Arg and FMOC- Har respectively, without the use of side chain protecting groups. This may be achieved by ensuring that post-coupling of the individual Arg or Har residue, the guanidino moiety is quantitatively protonated prior to any further coupling reactions and forms stable ion pair with the proton donor in organic solvent. This is preferably achieved by treating the resin bound peptide amide with an excess of the acidic coupling auxilliary BtOH or the like as described in more detail below in the experimental section. Another example of scavenging the charge of the guanidinhαm group is to use tetraphenyl borate salts of Fmoc- protected HAR for synthesis as set forth, in US 4,954,616.
The solid phase support or resin may be any support known in the art that is suitable for use in solid-phase synthesis. This definition of solid phase comprises that the peptide is bonded or linked via a functional linker or handle group to the solid phase or resin. Preferably the solid support is based on a polystyrene or polydimethylacrylamide polymer, as is customary in the art. According to the present invention, the peptide may be bonded via a suitable amino acid side chain, including e.g. the thiol moiety of a further cysteine residue of the peptide intended not to participate in the cyclization reaction, or may be bonded via the C-terminal α-carboxy group to a resin by means of e.g. an ether, thioetXier, ester, thioester or amide bond. Examples are solid supports comprising handle groups such as e.g. trityl, 2-chloro-trityl-, 4-methoxytrityl-, 'Rink amide' 4-(2',4'-dimethoxybenzyl- aminomethyl)-phenoxy-, Sieber resin (9-amino-6-phenylmethoxy-xanthen-), A- hydroxymethylphenoxyacteyl-, 4-bLydroxymethylbenzoic acid (the latter requiring attachement of the first amino acid by means of p-dimethylaminopyridine-catalysed esterification protocol than can result in racemisation of susceptible amino acids, e.g. Tip and in particular cysteine, see Atherton, E. et al., 1981, J. Chem. Soc. Chem. Coramun. , p.336 ff). Methods of providing thioester linkages to a resin are disclosed in detail and. are further referenced in WO 04/050686. Said reference also describes that thioester bonds are highly vulnerable to standard deprotection conditions used e.g. in Fmoc synthesis, and. how use of a substitute base may overcome this problem. However, in a preferred embodiment of the present invention, thioester linkages for bonding of the peptide moiety to the solid- phase, be it in a C-terminal or side chain born linkage, are specifically disclaimed since subject to transthioesterification side reaction under at least slightly basic pH. Thioester linkages are vulnerable to treatment with S-tert.butyl-sulphenyl protection group removing agents, in particular those of the thiol reducing type such as β-mercapto-ethanol in near- stochiometric amounts or beyond. But also with tertiary phosphines this may happen, setting free cysteinyl-, homo-cysteinyl, or generally residues with free thiol groups the latter which allowing further of intramolecular transthioesterification reaction with a solid- phase-anchoring thioester bond. However, the intramolecular reaction may be strongly modulated by aspects of spacial distance and sequence dependent, conformational restraints and hence applying the above disclaimer is dependent both on the type of S- tert.butyl-sulphenyl-group removing agent and the specific sequence of a given peptide. Preferably and optionally, where thioester linkages for bonding of the peptide moiety to the solid-phase are employed, the S-tert.butyl-sulphenyl protection group removing agent is a phosphine, more preferably a tris-(Cl-C8) alkyl-phosphine wherin the alkyl may be, independently, further substituted with halogeno or (Cl-4)alkoxy or (Cl-C4)ester. More preferably, the removing agent is a tris-(C2-C5)alkyl-phosphine wherein the alkyl may be further substituted,independently, with (Cl-C2)alkoxy.
Notably, according to the present invention, S-S-bond-comprising resin handles such as the HPDI bifunctional hydroxy and disulfide handle described in Brugidou, J. et al., Peptide Research (1994) 7:40-7 and Mery, J. et al, Int. J. Peptide and Protein Research (1993), 42: 44-52) are of course excluded from the scope of the present invention since not allowing of on-resin cyclization.
On-resin cyclization according to the present invention allows of avoiding the problems arising from intermolecular side reaction and the dilution techniques or catalyst-surface absorption techniques usually employed for this reason.
Rink amide, Sieber resin (Tetrahedron Lett. 1987, 28, 2107-2110) or similiar 9-amino- xanthenyl-type resins, PAL resins (Albericio et al., 1987, Int. J. Pept. Protein Research 30, 206-216) or the specially substituted trityl-amine derivatives according to Meisenbach et al., 1997, Chem. Letters , p. 1265 f.) are examples of linkage groups of a solid phase from which a Cα-carboxamid is generated or liberated upon cleavage of the peptide from the resin. In this sense solid phases giving rise to a carboxamid upon cleavage from resin, h>e it the carboxamid of a formerly acidic side chain or the C-terminus of the peptide, are termed amide-producing solid phases in the present context.
Preferably, the peptide is anchored to the solid phase by either an amide or ester bond via the C-terminus. More preferably, the solid phase is an acid-sensitive or acid-labile solid phase, even more preferably, it is an amide generating acid-labile solid-phase. Such acid- labile solid phases require at least 0.1% trifluoro acetic acid (TFA), more preferably at least 0.5% TFA in a polar aprotic solvent for cleavage from resin. Most preferably, the solid- phase is an acid-sensitive solid ptiase that is cleaved under weakly acidic conditions, that is 0.1 to 10% TFA in said solvent are sufficient to effect at least 90% cleavage efficiency upon incubation at room temperature up to 5 hours. Such highly acid-labile solid phase are e.g. 2-chlorotrityl resins, 4,4'-dirnethoxytrityl resin, the related, trityl-based phenylalcolhol resin such as e.g. Novasyn™ TGT derived from an conventional aminomethyl resin by acylation with Bayer's 4-carboxytrityl linker or a 4-methoxyphenyl, 4,4'-dimethoxyphenyl or 4-methyl-derivative of said linlcer, further Sieber resin, Rink amide resin or 4-(4- hydroxymethyl-3-methoxyphenoxy)-butyric acid (HMPB) resin, (4-methoxybenzhydryl-) or (4-methylbenzhydryl)-phenyl resins, the former said Sieber and Rink resin specifically giving rise to C-terminally amidated peptide upon acido lysis. Such acid-labile solid phases are particurlarly vulnerable to on-resin deprotection chemistries for side-chain protection groups and hence particular attention must be paid in these cases. In case of side chain anchoring via C-terminal cysteine residue to the handle group of a solid support, the linking bond must be a thioether or thioester bond. Further suitable residues for side-chain anchoring are carboxy groups of acidic side chains, hydroxy groups and in particular the ε-amino group of lysine. It goes without saying that in case of side chain anchoring, that the C-terminal free carboxygroup is generally to "be protected by esterification or amidation prior to carrying out the first coupling reaction, e.g. by using FMOC-Lys-carboxamid for linking reaction of the side chain amino function to the solid phase.
In a preferred embodiment, one S-alkyl-sulphenyl-protected cysteine, preferably one S- tert.butyl-sulphenyl protected cysteine is the C-terminal residue of the peptide and is bonded via the carboxy-terminus by means of an ester or amide bond to the solid phase, with the proviso, that said linking bond is not a benzylester moiety but preferably is an acid-labile resin that is cleaved under weakly acidic reaction conditions as defined above. A C-terminal cysteine is particularly prone to subject to racemisation in acidic conditions, e.g. upon cleavage and/or deprotection under strongly acidic condition.
Eventually disclaimed heterogenous catalysts for air-borne, oxidative cyclization are e.g. charcoal, which is incompatible Λvith use on a solid-phase. It may not be efficiently removed. Preferably, it relates to the absence of a catalytically effective or substantial amount of such heterogenous catalyst. Not using inappropriate catalyst when not required for the purposes of the present invention is a self-evident measure to the skilled artisan, though.
Coupling reagents for peptide synthesis are well-known in the art (see Bodanslcy, M. , Principles of Peptide Synthesis, 2nd ed. Springer Verlag Berlin/Heidelberg, 1993 ; esp. cf. discussion of role of coupling additivesauxilriari.es therein). Coupling reagents may be mixed anhydrides (e.g. T3P: propane phosphonic acid anhydride) or other acylating agents such as activated esters or acid halogenides (e.g. ICBF, isobutyl-chloroformiate) , or they may be carbodiimides (e.g. l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide), activated benzotriazin-derivatives (DEPBT: 3-(diethoxyphosphoryloxy)-l,2,3-benzotriazm-4(3H)- one) or uronium or phosphonium salt derivatives of benzotriazol. In view of best yield, short reaction time and protection against racemization during chaing elongation, more preferred is that the coupling reagent is selected from the group consisting of uronium salts and phosphonium salts of the benzotriazol capable of activating a free carboxylic acid function along with that the reaction is carried out in the presence of a base. Suitable and likewise preferred examples of such uronium or phosphonium. coupling salts are e.g. HBTU (0-lH-benzotriazole-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), BOP (benzotriazole- 1 -yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate) , PyB OP (B enzotriazol- 1 -yl-oxy-tripyrrolidinopho sphonium hexafluorophosphate), PyAOP, HCTU (O-ClH-ό-chloro-benzotriazole-l-yrj-U ,3, 3- tetramethyluronium hexafluorophosphate), TCTU (O-lH-6-chlorobenzotriazole-l-yl)- 1,1,3,3-tetramethyluronium tetrafluoroborate), HATU (O-(7-azabenzotriazol-l-yl)-l,l,3,3- tetramethyluronium hexafluorophosphate), TATU (O-(7-azabenzotriazol-l-yl)-l,l ,3,3- tetramethyluronium tetrafluoroborate), TOTU (O- [cyano(ethoxycarbonyl)methyleneamino]-N,N,N',N"-tetramethyluronium tetrafluoroborate), HAPyU (O-(benzotriazol- 1 -yl)oxybis-(pyrrolidino)-uronium hexafluorophosphate.
Preferably, when using DEPBT or the like, uronium or phosphonium salt reagents, a further or second weak base reagent is needed for carrying out the coupling step. This is matched by base whose conjugated acid has a pKa value of from pKa 7.5 to 15, more preferably of from pKa 7.5 to 10, with the exclusion of an α-amino function of a peptide or amino acid or amino acid derivative, and which base preferably is a tertiary, sterically hindered amine. Examples of such and further preferred are Hϋnig-base ( N,N- diisopropylethylamine), N,N'-dialkylaniline, 2,4,6-trialkylpyridine or N-alkyl-rαorpholine with the alkyl being straight or branched C1-C4 alkyl, more preferably it is N- methylmorphorine or collidine (2,4,6-trimethylpyridine), most preferably it is collldine.
The use of coupling additives, in particular of coupling additives of the benzotriazol type, is also known (see Bodansky, supra). Their use is particularly preferred when using the highly activating, afore said uronium or phosphonium salt coupling reagents. Hence it is further preferred that the coupling reagent additive is a nucleophilic hydroxy compound capable of forming activated esters, more preferably having an acidic, nucleophilic N- hydroxy function wherein N is imide or is N-acyl or N-aryl substituted triazeno, most preferably the coupling additive is a N-hydroxy-benzotriazol derivative (or 1 -hydroxy- benzotriazol derivative) or is an N-hydroxy-benzotriazine derivative. Such coupling additive N-hydroxy compounds have been described in large and wide in WO 94/07910 and EP -410 182 and whose respective disclosure is incorporated by reference hereto. Examples are e.g. Kf-hydroxy-succinimide, N-hydroxy-3,4-dihydro-4-oxo-l,2,3- benzotriazine (HOOBt), l-hydroxy-7-azabenzotriazole (HOAt) and NT-hydroxy- benzotriazole (HOBt). N-hydroxy-benzotriazine derivatives are particularly preferred, in a most preferred embodiment, the coupling reagent additive is hydroxy-3,4-dihyclro-4-oxo- 1,2,3-benzotriazine.
Ammonium salt compounds of coupling additives are known and their use in coupling chemistry has been described, for instance in US4806641.
In a further particularly preferred embodiment, the uronium or phosphonium salt coupling reagent is an uronium salt reagent and preferably is HCTU, TCTU or HBTU and even more preferably is used in the reaction in combination with N-hydroxy-3,4-dihydro-4-oxo- 1,2,3-benzotriazine or a salt thereof. This embodiment is mainly preferred for use in chain elongation step of peptide synthesis after removal of the base-labile Nα-protection group, but may as well be used for lactamization reaction during side-chain cyclization.
In the context of the present invention, it is to be noted that HCTU and TCTU are defined as to be encompassed by the term 'uronium salt reagent' despite that these compounds and possible analogues have been shown to comprise an isonitroso moiety rather than an uronium moiety by means of crystal structure analysis (O. Marder, Y. Shvo, and F. Albericio "HCTU and TCTU: New Coupling Reagents: Development and Industrial Applications", Poster, Presentation Gordon Conference February 2002), an IM-amidino substituent on the heterocyclic core giving rise to a guanidium structure instead, hi the present context, such class of compounds is termed 'guanidinium-type sαbclass' of uronium salt reagents according to the present invention.
In a further particularly preferred embodiment, the coupling reagent is a phosphonium salt of the benzotriazol such as e.g. BOP, PyBOP or PyAOP. Deprotection of the base labile Na may be carried out as routinely done in the art, e.g. with 20% piperidine in N-methyl morpholine when using standard Fmoc chemistry. Most widely, Fmoc or Boc protection chemistry for the N-terminus is routinely applied in solid phase synthesis but further optional Na protection chemistries are known in the art and can be applied where not interfering with the present invention, that is to devise disullide-borne peptide cyclization of the resin-conjugated peptide.
The S-alkyl-sulphenyl protecing groups protecting thiol groups of cysteine or homocysteine residues, as is shown in formula II, are removed according to the present invention by a reagent that typically is capable of removing, preferably substantially removing, the S-tert.butyl-sulphenyl-protection group from such residue. Removal of S- tert.butyl-sulphenyl protection groups from e.g. cysteine accomplished by means of reaction with tertiary phosphines has been described, for instance by using tributylphosphine (Atherton et al., 1985, J. Chem. Soc, Perkin I. 2057) and triethylphosphine (Huang et al, 1997, Int. J. Pept. Protein Res. 48, 290). The tert- butylsulphenyl group is also cleaved in an orthogonal fashion by means of thiol reagents such as e.g. β-mercapto-ethanol or dithio-threitol (DTT) as an option to using tertiary phosphines (Huang et al.,1997 Int. J. Pept. Protein Res. 48, 290; Rietmann et al., 1985, Reel. Trav. Chim. Pays-Bas, 1141). Preferably, the tertiary phosphine is triphenylphospine or is an (C1-C4) alkylated or (Cl-C4)alkoxylated triphenylphosphine, such as e.g. tri-(p- methoxyphenyl)-phosphine or even more preferably is a trialkylphosphine wherein the alkyl may be the same or different , and wherein each alkyl is a Cl to C7 alkyl, preferably Cl to C4 alkyl, and maybe branched or linear alkyl. Preferably, the alkyl is linear. Examples are methyl, ethyl, propyl, i-propyl, n-butyl, i-butyl. Tri-n-butyl-phosphιine and tri-ethylphosphine are particularly preferred. The alkyl may be optionally further substituted with halogeno, (Cl-C4)alkoxy such as e.g. and preferably methoxy or ethoxy, or may be further substituted where amenable with the solvent system, carboxy or is, preferably, unsubstituted. Surprisingly, in one preferred embodiment according to the present invention, it has unexpectedly been found that disulfide cleavage by means of phosphines may also be used with the acid-labile resins cleavable in weakly acidic reaction conditions such as Sieber or 2-chloro-trityl (CTC) resin, for instance. It is also often overlooked that thiol reagens reduce and hence cleave disulfides "by forming disulfide products themselves. Preferably, such thiol reagent is selected from the group consisting of erythro-2,3-dihydroxy-l,4-butanedifhiol (or named meso-1,4- Dithioerythritol or DTE for short ), DL~threo-2,3-dihydroxy-l,4-butanedithiol (or named rac-l,4-Dithiothreitol or DTT for short), L-threo-2,3-dihydroxy-l,4-butaneditliiol, D- threo-2,3-dihydroxy-l,4-butanedithiol and mixtures thereof. Mixtures may comprise DTE and DTT, either in its racemic form or as an optically active preparation of DTT. More preferably, the thiol reagent is DTT which means D-,L- or any racemic or non-racemic mixture thereof. - DTT and DTE are also known as Cleland's reagent and Cleland's other reagent, respectively (Cleland, W. , Biochemistry 3,480-482,1964). Whereas in case of DTT, intramolecular ring closure is strongly favored, making it a stronger disulfide reducing agent and notably preventing formation of stable intermolecular disulfide adducts with DTT, in case of β-mercaptoethanol, any intermolecular reaction product, by way of disulfide exchange reaction, is feasible. Further newly formed disulfides may undergo further exchange reaction. The use of thiol reagents, most oftenly simple thiol reagents such as 2-mercaptoeth.anol, apparently owes to the fear of side reactions such as e.g. leakage from resin when using strongly nucleophilic tertiary phosphine reagents. By using DTT and the like, the inherent disadvantages of mono-thiol reagents maybe avoided.
Cyclization is carried out according to the present invention in the presence of a first weak base in a polar, aprotic organic solvent in the presence of air and/or oxygen but notably in the absence of a heterogenous, rate-accelerating catalyst. Still then, and without precedent, the cyclization step, due to the method of the present invention, is remarkably efficient and requires only about 0.5 to 2 hours reaction time, allowing of literally quantitative, complete conversion of educt to the desired product under very mild reaction conditions (ambient temperature typically, expedient temperature range being 1O0C to 8O0C though reflux temperature of solvent must be taken into account of course). Conversion is complete. This is an outstanding achievement and has not yet been achieved in disulfid-bonding driven cyclization of peptide, nor have such simple, mild and rapid cyclization reaction conditions been devised earlier. No tedious mixing and separation problems for a heterogenous catalyst arise ever. Still, the reaction rate completely parallels that of the catalyst-borne reaction of the prior art. Due to the straightforward course of reaction, formation of side products is almost entirely avoided. Suitable polar, aprotic solvents are e.g. acetonitril, dimethylformamide, dichloxomethane, N-methyl-pyrrolidone, tetrahydrofurane. In contrast to water, such solvent usαally may not physically dissolve relevant amounts of oxygen to supply the oxidative formation of disulfide bonds as has been described for aequeous catalyst systems before.
Accordingly, he supply of air, air/oxygen or pure oxygen must be paid attention to. Air/oxgen maybe supplied by thorough stirring, vortexing, special design of propellers used for stirring, gas sparging into the liquid. The gas may be air or pure oxgen or air enriched with oxgen which is vented or sparged into the reaction liquid. In on_e particularly preferred embodiment, large or surface areas of the bottom and/or walls of the reactor vessel are puntctured as to allow of sparging gas into the liquid, under thorough stirring. More preferably, the reaction vessel comprises a fritted bottom or a fritted section of least 50% surface area of the total surface of the bottom, as to allow of simultaneous stirring and venting by upsurging, bubbling air vented into the reaction liquid through that bottom.
The first weak base reagent is a weak base whose conjugated acid has a pKa v^alue of from pKa 7.5 to 15, more preferably of from pKa 8 to 10, preferably, it is a tertiary;, sterically hindered amine. Examples of such and further preferred are Hϋnig-Base (N5N"- diisopropylethylarαine), N,N-dialkyl-aniline, 2,4,6-trialkylpyrididine or N-allcyl- morpholiiie with the alkly being straight or branched C1-C4 alkyl such as methyl, ethyl, n- propyl, i-propyl, n-butyl, most preferably it is N-methylmorpholine, collidine (2,4,6- trimethylpyridine) or Hϋnig-Base.
Preferably the prior removal of the disulfide-bonded protection group according to the present invention, notably the removal of the S-tert.butyl-sulphenyl group is effected in the presence of a first -weak base reagent, for avoiding any risk of leakage from ttaie resin by minor acidolysis, tliat is at a pH of from 7.5 to 12, more preferably of from 8 "to 11. Optionally, by using polar aprotic solvents such as THF or acteonitril that are freely miscible with water, basic salts such as e.g. sodium acetate in aequeous solution may be used for that purpose. This embodiment is particularly preferred when using tertiary phosphines for said, disulfide group cleavage or removal step. By combining a suitable oxygen supply concomittant with such disulfide protection group removal, it may be possible in another embodiment of the present invention, e.g. when using polar, aprotic organic solvent along with oxygen supply in the presence of a tertiary amine and when using tertiary plαosphine for deprotection that is inert to oxygen, to carry out both disulfide deprotection and cyclization not only in a one-pot reaction but even as a single reaction step.
Due to the fact that the present method allows of on-resin cyclization, it further does not require tedious and yield decreasing strong dilution of peptide for favoring intra over intermolecular cyclization as previously required in most methods described in the prior art. The on-resin operation mode of the invention allows of quick and efficient intra-molecular cyclization only, giving no chance of dimerization at all.
In a further preferred embodiment, the peptide is the peptide of formula I or II. The term protection group is to be construed as being protection group for a gi~ven side chain functionality or specific side chain which protection group is compliant with being used in standard tert-bixtyloxycarbonyl (Boc) or 9-fluorenylmethoxycarbonyl (Fmoc) solid phase peptide synthesis. Such protection groups and the use of specific protection groups for specific side chain functionalities are well-known in the art (s. Chan et al., ed., supra; Bodansky et al. supra).
A side chain group Rl (o) for instance it not to be construed in the way as to refer to a single type of optionally protected amino acid side chain; each residue Rl(I), Rl(2)...may be unique or may be the same as at least one other residue. The same applies of course to radicals R2(x), R3(q).
Given the multitude of possilbe substructure, it is to be noted that the peptide of formulas I and II may also comprise well-known peptide backbone modifications that are commonly employed in peptide synthesis: cyclic amino acids such as D- or L-Pro, intermittent non- peptide moieties linking two peptidyl segments and being e.g. hairpin or β-turn mimetics or in particular backbone-modified dipeptidyl segments used in synthesis e.g. for introducing amide protected Asp-Gly(Hmb) segments avoiding aspartimide formation (Packman et al. 1995, Tetrahedron Lett. 36, 7523) or peptidomimetic, non-amide bonded dimeric segments of amino acids analogs having a backbone segments such as -CO-CH2- or CH2-NH2- instead of a peptide bond (a review of useful peptidomimetic segments can be found, e.g. in Morley, J., Trends Pharm. Sci. (1980), pp.463-468).
Preferably, the two cysteines that are going to be disulfide-connected in cyclization are spaced apart by at least two amino acid residues (or the like). A spacing of i+3 is typical of an α-lielical peptide conformation and allows of optimal,spacial juxtaposition for disulfide bonding. In this way, cyclization is facilitated. Below, the constraint exercised by the backbone in view of the possible, more stable conformations is rendering cyclization more difficult. However, it is to be noted that the incorporation of pseudo-prolines as helix breakers or of D-amino acids as inducers of beta-turn conformations in the spacer segment of the peptide moiety, that is as one of the amino acids encompassing a radical R2(x), is strongly modulating this simple rule, which is highly structure dependent accordingly.
In one further embodiment, it may be possible to synthesize a peptide on a solid phase not by permanent, covalent attachment of the peptidyl moiety to a solid-phase but by non- covalent, reversible attachement to the solid-phase by means of a stable metal chelate complex (pressrelease October/November 2004 made by Lonza AG, Basel, Switzerland jointly with AplaGen GmbH, Baesweiler, Germany, October 20O4), similiar to the hexa- His tag technology employed in protein purification since long. Such non-covalent solid- phase linkage or similiar, future embodiments are encompassed by the present invention as well and the preferred modes of operating the present invention set forth above and in the claims below apply to this embodiment, substituting the afore mentioned linkage or bonding to the resin or handle with the non-covalent bonding feature of said present embodiment..
A further object of the present invention are the respective, solid-phase borne peptides or solid-phase-peptide conjugates, respectively. The relevant definitions given above and beloΛV apply likewise to such object, alone or in combination.
Accordingly said further object of the present invention is a peptide of formula I or II,
Figure imgf000016_0001
or
Figure imgf000016_0002
II
wherein m,n=l-15, preferably m,n = 1 or 2, wherein Y=EE or Y is first a protection group, preferably Y is an non-base labile protection group, o,x,q each separately is 0-200 and wherein Rl (o), R2(x), R3(q) each, independently, is a side chain of an amino acid selected from the group consisting of natural amino acids including cyclic amino acids, non-natural amino acids including cyclic amino acids or non-amide bonded dipeptidyl segements, non-natural derivatives of natural amino acids or analogues thereof and wherein said amino acid chain may each further comprise a second protection group that may be the same or different for individual side chains, and wherein A is a resin or resin handle or wherein optionally an individual R2(x) or R3(q) radical is linked to a resin or resin handle with the proviso that then A is selected from the group comprising OH, NH2, NR' iH or IS[R' ΪR'2 with R'lj2 being Cl to C4 alkyl, and wherein RlO, Rl 1 each are alkyl which may be further substituted with aryl, aryloxy, alkoxy, halogenated variants thereof or halogeno and may be the same or different.
Preferably, x = 2-200. More preferably, o,x,q each separately is 1-100, preferably 2-50, or wherein x is 2-100, preferably x is 3-50. Again more preferably, q=0 and more preferbly in this context further o is 0-50 and wherein x is 2-100, preferably wherein x is 2-50.
Experiments
The overall synthetic strategy is set forth in table I underneath:
Table I
3x, 15 min
Peptide x for Har), lengthening , NMP/DCM
Figure imgf000018_0001
Boc-Gly-Cys(S-tBu)-Har-Gly-Asp(tBu)-Trp(Boc)-Pro-Cys(S-tBu)-NH-
PBu3 mediated cleavage cyclization on support
Boc-Gly-Cys-Har-Gly-Asp(tBu)-Trp(Boc)-Pro-Cys-NH — T Sieber
Cleavage from the resin Global deprotection
Gly-Cys-Har-Gly-Asp-Trp-Pro-Cys-NH2
I I
1. 1 FMOC solid phase synthesis of linear peptide Fmoc-Gly- Asp(tBuVTrp(Boc)-Pro- CvsfS-tBuVSieber
Synthesis of FMOC-Cys(S-tBu)-OH has been described "before (Rietman et al., 1994, Synth. Commun. 24, p. 1323 f). Sieber resin was a Novabiochem™ product of 100-200 mesh. (US Bureau of Standards mesh sizing), the matrix material being divinylbenzene- crosslinked polystyrene, and was purchased from Calbiochem-Novabiochem (belonging to EMD Biosciences, California/U.S.A.). All FMOC amino acids, including FMOC-Cys(S- tBu)-OH (cat. No. B-1530) were purchased from Bachem AG (Bubendorf, Switzerland).
Loading of resin was at 0.52 mmol/g and of a total of 10 g Sieber resin. Coupling time for loading was twice the standard coupling time, namely 60 min. in total. Couplings were conducted with 2 eq. each of respective amino acid in the presence of leq. each of 6- chloro-HOBt, TCTU, Hϋnig-Base (Disopropylethylamiae), in dichloromethane. Washes were with N-methyl-pyrrolidone (NMP).
FMOC deprotection was done by 3 cycles of 15 min. 10*% piperidine in N-methyl- pyrrolidone; efficiency of cleavage and completion of synthesis was analysed by Ninhydrin reaction and reverse phase HPLC, respectively.
1.2 Elongation of peptide from 1.1 to Boc-Gly-CvsfS-tBiiVHar-Glv-AspftBuVTrpfBoc)- Pro-Cvs(S-tBu)-Sieber
The coupling of the FMOC-Har residue (Bachem, Burgendforf, Switzerland) took place in the presence of 1 eq. HOBt (for keeping the Guanidino group protonated) per eq. amino acid; the FMOC amino acid was preincubated with HOBt and diisopopyl-carbodiimid in NMP and was then mixed with the resin. Har coupling took 180 min. (other aa: 30 min.) followed by a second cycle with replenished reagents of about 60 min. In this way, standard 99.8 % coupling efficiency as for the other residues could be matched. FMOC cleavage took place as before. Notably, after FMOC cleavage und subsequent NMP washes, repeated washing with HOBt was done to prevent further swelling of the resin
1.3 Deprotection of protected (S-tBu)-cysteines with Bu^P
The resin product of step 1.2 was suspended and washed three times in tetrahydrofurane (THF). The reaction was carried out for 1 h at room temperature with 50eq. tributylphosphine made up as 19%(v/v) PBu3 /77%(v/v) THF /4%(v/v) saturated aequeous solution of sodium acetate; precipitating salt was filtered off prior to use . Reaction proceded uniformly to give one dominant product peak. The yield was determined by reverse phase HPLC and was found to amount to 98.9%correct product.
1 .4 Cvclization to yield Boc-Glv-cvcfofCvs-Har-Gly-Asp('tBuVTrp(Boc)-Pro-Cvs)-Sieber
The swollen peptide-resin conjugate from exp. 1.3 was washed three times in NMP. Cyclization was done by incubating the resin for 1 h at room temperature with 6% DIEA (Hϋnig-Base) in NMP; reaction was carried out in a vertical glass vessel which comprised a horizontally bisecting, sealed-in G3 (16-40 μm) glass frit in its lower portion. The glass frit or fritted plate was vented with air from below, allowing of air bubbling across the entire cross-section of the solvent-covered reactant space above the frit in which the resin was floating by the bubbling air from underneath. A strictly pure, uniform product is obtained, no distinct or shattered sideproducts do show off after this reaction step. The conversion to product was 100%, as determined independently by both reverse phase HPLC and LC-MS. RP-HPLC was carried out on a Hypersil-Keystone™ Betabasic (Thermo Electron Corp., Waltham Mass./U.S.A.) Cl 8 150x4.6 mm column, with an injection volume of 15 μl and detection at 262nm at a column temperature of 350C. Gradient run is
Time Acetonitrile (0.1 %TFA)/Water (0.1 %TFA) O 60 40
5 97 3 16 97 3
17 60 40
1 .6 Global deprotection
Global deprotection is prepared by swelling the resin three times in dichloromethane (DCM). Cleavage reaction phase mixture is prepared as to be made up from 86.5% TFA (785eq.) 4.5% Thioanisol (36.5eq.) 3 % Phenol (32.4 eq.) 3% DCM (38 Eq.)
3% H2O (178 Eq.)
Reaction takes place at 150C for 2 h on an slowly rotating orbital shaking device. Reaction is terminated and product is precipitated, after filtering off the resin, by dropwise addition of tert.butyric acid methyl ester. The product is a uniform peak; no major sideproduct can be detected.- the above conditons of global deprotection have been tested on a control and found not to affect preformed disulfide bridges in peptides.
2. Deprotection of protected (S-tBu)-cysteines with DTT
As an option to the deprotection step in 1.3, deprotection is carried out with DTT instead of phosphin essentially as described there. DTT is either rac- or L-DTT, obtainable from Biosynth AG/Switzerland. The resin product of step 1.2 was suspended and washed three times in dimethylformamide (DMF). 50 eq. of DTT were used, made up as DMF/DTT (1 :1) and the reaction time was extended to 3-5 hours at room temperature. Subsequently, the peptide-resin was treated exactly as described in section 1.4-1.6 above. Yields obtained perfectly matched that of 1.6, with similar purity.
3. Cvclisation of Boc-D-Phe-Cvs(S-tBu) -Tyr(tBu)-D-Trp(Pb:Q-LysfBocyVal-Cys(S-tBuV TrpfPbf)-Sieber
Vapreotide, a Somatostatin peptidagonist, is synthesized essentially as described above in section 1.1. Further processing is carried out essentially as described in sections 1.2-1.6, providing the deprotected Vapreotide-carboxamide in excellent yield and purity. Optionally, deprotection according to section 2. is carried out, likewise with very good result. 4. Cvclisation of BOC-LVS(BOC)-CvS(S-IBu)-ASn-ThT(TIt)-AIa-ThXfTn)-CvSfS-IBu)-AIa-
Thr(Trt)-Gln-Arg(Pbf)-Leu-Ala-Asn-Phe-Leu-Val-His-Ser(Trt)-Ser(Trt)-Asn-Asn-Phe-
Glv-Pro-Ile-Leu-Pro-Pro-Thr(Trt)-Asn-Val-Glv-Ser(Trt)-Asn-Thr(Trt)-Tyr-Sieber Pramlintide peptide, a 37-mer, is synthesized and cyclized essentially as described above in sections 1.1-1.6. As compared to the yield of linear peptide, cyclization itself is quantitative. However, full length C to N-terminal linear synthesis give mediocre yield, due to several difficult individual coupling steps.
5. Cvclization of Fmoc-Cvs(S-fBu)-Asn-Thr(TrtV Ala-Thr(Trt)-C vs(S-tBu)-Ala-Thr(Trt)- Gln-Arg(Pbf)-Leu-Ala-Asn-Phe-Leu-Val-His-Ser(Trt)-Ser(Trt)-Asn-Asn-Phe-Glv-Pro-Ile- Leu-Pro-Pro-Thr(Trt)-Asn-Val-Glv-Ser(Trt)-Asn-Thr(Trt)-Tyr-Pvinl<:
Synthesis is essentially carried out as described in section 4. above, except that the last Lys residue is added after cyclization reaction in an additional coupling cycle, that synthesis is carried out on a Rink amide resin and that prior to global deprotection, cleavage is carried out under weakly or mildly acidic condition: Cleavage from resin is achieved with 3 cycles of 15 min. each at 150C, 2% (w/w) TFA, 1% (w/w) triethylsilane (TES) in dichloromethane. The reaction is stirred by nitrogen bubbling. After each cycle, cleavage reaction is directly quenched by pouring the whole reaction broth into dilute pyridin
(pyridine/ethanol 1 :9 (v/v)). Resin is then removed by filtration with a frit. Filtrates are pooled and concentrated under vacuo (RotaVap), and washed with DCM.
6. Cvclization of Boc-LvsfBoc)-Cys(S-tBu)-Asn-Thr(Trt)-Ala-Thr(Trt)-Cvs(S-tBu)-Ala- (\ι/Me'Mepro)Thr-2-CTC
Synthesis and cyclization is essentially carried out as described in sections 1.1-1.5 above,except that 2-chlorotrityl-polystyrene resin (CBL Patras, Greece) is used as a solid phase and that the DTT method according to section 2 is used instead of 1.3/phosphine method. Further, cleavage under mildly acid condition without side chain deprotection is used, essentially as described in section 5. Good yields are obtained. Fragment synthesis serves as an optional route to Pramlintide synthesis: the cylized, bridging-cystine comprising but still protected peptide is then subjected to conventional fragment coupling technique with a C-terminal, residual fragment of Pramlintide using standard peptide coupling chemistry with TCTU wherein the C-terminal, protected fragment is harbored either on solid-phase or, preferably, in liquid phase, too.

Claims

Claims
1. Method of peptide synthesis, comprising the steps of
a. synthesizing a peptide linked to a solid phase which peptide comprises at least two residues of a cysteine or a homo-cysteine, which cysteines are protected in their side ctiain each by a S-alkyl-sulphenyl protection group, wherein the alkyl may be further substituted with aryl, aryloxy, alkoxy, halogenated variants thereof or halogeno, and wherein the two protection groups may be the same or different, preferably they are protected in their side chain each by a S-tert.butyl-sulphenyl group, and
b. reacting the peptide with, a S-tert.Butyl-sulphenyl-protection group removing reagent, preferably reacting it with a tertiary phosphine, and
c. cyclizing the peptide by means of disulfide bond formation in the presence of air and/or oxygen.
2. Method according to claim 1, characterized in that said cysteines are spaced apart by at least 3 residues.
3. Method according to claim 1, characterized in that the solid phase resin is an acid- labile resin, preferably one that is selected from the group consisting of 2-chloro- trityl, HMPB, Sieber or Rink amide resin which are resins labile under weakly acidic condition of from 0.1% (v/v) to 10% (v/v) trifluoroacetic acid in polar, aprotic solvent and wherein, preferably, the last C-terminal residue is a cysteine or homo-cysteine and is C-terminally linked to the resin which resin, more preferably, is a Sieber or Rink amide resin.
4. Method according to claim 1 , characterized in that the peptide has at least one further side chain protection group which is protection group is not a S-alkyl- sulphenyl protection group including differently protected further cysteine or homo-cysteine residues.
5. Method according to claim 1 or 3, characterized in that the homo-cysteine comprises in its side chain 2-15 methylene groups and one thiol group.
6. Method according to claiml or 6, characterized in that the removal of the S-all<yl- sulphenyl groups is accomplished by reacting the peptide with a trialkylphosptiine.
7. Method according to claiml , characterized in that the peptide is cyclized in the presence of a weak base in a polar, aprotic solvent.
8. Method according to claim 1 or 7, characterized in that the linkage of the peptide to the solid phase is acid labile, preferably labile in 60% TFA in dichloro-methane at room temperature.
9. Method according to claim 5, characterized in that the resin is a Sieber resin.
10. Method according to claim 1, characterized in that in a subsequent step, the peptide is cleaved off from the resin, preferably under condtions of global deprotection.
11. Method according to claim 1 or 3, characterized in that the linkage of the peptidyl moiety to the solid phase resin is not a thioester or disulfide bond linkage.
12. Peptide of formula I or II,
Figure imgf000025_0001
or
Figure imgf000025_0002
II
wherein m,n=l-l 5 , preferably m,n = 1 or 2, wherein Y=H or Y is first a protection group, preferably Y is an non-base labile protection group, o,x,q eacli separately is 0-200 and wherein Rl (o), R2(x), R3(q) each, independently, is a side chain of an amino acid selected from the group consisting of natural amino acids including cyclic amino acids, non-natural amino acids including cyclic amino acids or non- amide bonded dipeptidyl segements, non-natural derivatives of natural amino acids or analogues thereof and wherein said amino acid chain may each fixrther comprise a second protection group that may be the same or different for individual side chains, and wherein A is a resin or resin handle or wherein optionally an individual R2(x) or R3(q) radical is linked to a resin or resin handle with the proviso that then A is selected from, the group comprising OH, NH2, NEV1H or NR' χRJ2 with R'1;2 being Cl to C4 alkyl, and wherein RlO, Rl 1 each are alkyl which may be further substituted with aryl, aryloxy, alkoxy, halogenated variants thereof or halogerxo and may be the same or different.
13. Peptide according to claim 12, characterized in that x = 2-200.
14. Peptide according to claim 12 or 13, characterized in that o,x,q each separately is 1-100, preferably 2-50, or wherein x is 2-100, preferably x is 3-50.
15. Peptide according to claim 12-14, characterized in that q=0.
16. Peptide according to claim 15, characterized in that o is 0-50 and wherein x is 2- 100, preferably wherein x is 2-50.
PCT/EP2005/011476 2004-10-16 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis WO2006045603A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP05810158A EP1807442A1 (en) 2004-10-26 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis
AU2005298840A AU2005298840B2 (en) 2004-10-26 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis
US11/666,171 US20080108790A1 (en) 2004-10-16 2005-10-26 S-Alkyl-Sulphenyl Protection Groups in Solid-Phase Synthesis
BRPI0518390-1A BRPI0518390A2 (en) 2004-10-26 2005-10-26 Peptide Synthesis Method and Peptide
JP2007538330A JP2008529969A (en) 2004-10-26 2005-10-26 S-alkyl-sulfenyl protecting group in solid phase synthesis
CA002584906A CA2584906A1 (en) 2004-10-26 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis
CN2005800365194A CN101072791B (en) 2004-10-26 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis
IL182751A IL182751A0 (en) 2004-10-26 2007-04-23 S-alkyl-sulphenyl protection groups in solid-phase synthesis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04025395.7 2004-10-16
EP04025395 2004-10-26

Publications (1)

Publication Number Publication Date
WO2006045603A1 true WO2006045603A1 (en) 2006-05-04

Family

ID=36090794

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/011476 WO2006045603A1 (en) 2004-10-16 2005-10-26 S-alkyl-sulphenyl protection groups in solid-phase synthesis

Country Status (11)

Country Link
US (1) US20080108790A1 (en)
EP (1) EP1807442A1 (en)
JP (1) JP2008529969A (en)
KR (1) KR20070083815A (en)
CN (1) CN101072791B (en)
AU (1) AU2005298840B2 (en)
BR (1) BRPI0518390A2 (en)
CA (1) CA2584906A1 (en)
IL (1) IL182751A0 (en)
TW (1) TW200637872A (en)
WO (1) WO2006045603A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008080845A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Methods for the synthesis of cyclic peptides
WO2009003666A1 (en) * 2007-06-29 2009-01-08 Lonza Ag Process for the production of pramlintide
EP2334314A1 (en) * 2008-09-03 2011-06-22 ScinoPharm Taiwan, Ltd. Process for the preparation of pramlintide
CN102180943A (en) * 2010-12-16 2011-09-14 深圳市健元医药科技有限公司 Production process of polypeptide medicament for assisting to reduce blood sugar
CN112111001A (en) * 2019-06-19 2020-12-22 翰宇药业(武汉)有限公司 Method for synthesizing thymosin T alpha-1

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2981352B1 (en) * 2011-10-17 2015-07-03 Centre Nat Rech Scient PROCESS FOR PROTEIN SYNTHESIS
CN102875639A (en) * 2012-09-26 2013-01-16 深圳翰宇药业股份有限公司 Solid-phase synthetic method of peptide and peptide synthesized by same

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010146A1 (en) * 1991-11-19 1993-05-27 Amylin Pharmaceuticals, Inc. Amylin agonist peptides and uses therefor
US6087334A (en) * 1998-08-21 2000-07-11 Amylin Pharmaceuticals, Inc. Anti-diabetic peptides
EP1164143A1 (en) * 2000-05-23 2001-12-19 Institute Of Nuclear Energy Research, Taiwan, R.O.C. Process for preparing octreotide and derivatives thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5889146A (en) * 1997-11-26 1999-03-30 Institute Of Nuclear Energy Research Method for synthesis of bifunctional chelating agents-peptides
BR0113091A (en) * 2000-11-27 2003-07-08 Rmf Dictagene Sa Process for the multiplication of chemically synthesized polypeptides and process for the preparation of biologically active proteins

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010146A1 (en) * 1991-11-19 1993-05-27 Amylin Pharmaceuticals, Inc. Amylin agonist peptides and uses therefor
US6087334A (en) * 1998-08-21 2000-07-11 Amylin Pharmaceuticals, Inc. Anti-diabetic peptides
EP1164143A1 (en) * 2000-05-23 2001-12-19 Institute Of Nuclear Energy Research, Taiwan, R.O.C. Process for preparing octreotide and derivatives thereof

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008080845A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Methods for the synthesis of cyclic peptides
JP2010514728A (en) * 2006-12-29 2010-05-06 エフ.ホフマン−ラ ロシュ アーゲー Synthesis method of cyclic peptide
WO2009003666A1 (en) * 2007-06-29 2009-01-08 Lonza Ag Process for the production of pramlintide
EP2334314A1 (en) * 2008-09-03 2011-06-22 ScinoPharm Taiwan, Ltd. Process for the preparation of pramlintide
EP2334314A4 (en) * 2008-09-03 2012-03-21 Scinopharm Taiwan Ltd Process for the preparation of pramlintide
CN102180943A (en) * 2010-12-16 2011-09-14 深圳市健元医药科技有限公司 Production process of polypeptide medicament for assisting to reduce blood sugar
CN112111001A (en) * 2019-06-19 2020-12-22 翰宇药业(武汉)有限公司 Method for synthesizing thymosin T alpha-1
WO2020252883A1 (en) * 2019-06-19 2020-12-24 深圳翰宇药业股份有限公司 SYNTHESIS METHOD FOR THYMOSIN Tα-1
CN112111001B (en) * 2019-06-19 2021-10-29 翰宇药业(武汉)有限公司 Method for synthesizing thymosin T alpha-1

Also Published As

Publication number Publication date
KR20070083815A (en) 2007-08-24
TW200637872A (en) 2006-11-01
CA2584906A1 (en) 2006-05-04
EP1807442A1 (en) 2007-07-18
CN101072791B (en) 2011-01-19
JP2008529969A (en) 2008-08-07
AU2005298840A1 (en) 2006-05-04
BRPI0518390A2 (en) 2008-11-18
CN101072791A (en) 2007-11-14
AU2005298840B2 (en) 2010-09-16
IL182751A0 (en) 2007-07-24
US20080108790A1 (en) 2008-05-08

Similar Documents

Publication Publication Date Title
ES2364982T3 (en) SYNTHESIS OF SIMULAR PEPTIDES TO GLUCAGON.
EP0212432B1 (en) Calcitonin gene related peptide derivatives
AU2005298840B2 (en) S-alkyl-sulphenyl protection groups in solid-phase synthesis
EP1794180B1 (en) Peptide cyclisation
AU2020334993B2 (en) Methods of making incretin analogs
CN111566123A (en) Process for preparing glucagon-like peptide
ES2938734T3 (en) Procedure for the manufacture of analogues of GLP-1
RU2361876C2 (en) Peptide vectors
US6346601B1 (en) Procedure for obtaining the somatostatin analog, octreotide
US6673769B2 (en) Lanthionine bridged peptides
KR101222774B1 (en) On-resin peptide cyclization
KiSFALUDY et al. Pentagastrin Analogs Containing α-Aminooxy Acids, I Synthesis of Analogs Substituted at the N-Terminus
EA047599B1 (en) METHODS OF OBTAINING INCRETINN ANALOGUES
Elmore Peptide synthesis
Wünsch SECOND JOSEF RUDINGER MEMORIAL LECTURE KAISER-WILHELM-/MAX-PLANCK-GESELLSCHAFT. RESEARCH IN PEPTIDE CHEMISTRY FROM 1921 TO 1991?

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BW BY BZ CA CH CN CO CR CU CZ DK DM DZ EC EE EG ES FI GB GD GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV LY MD MG MK MN MW MX MZ NA NG NO NZ OM PG PH PL PT RO RU SC SD SG SK SL SM SY TJ TM TN TR TT TZ UG US UZ VC VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SZ TZ UG ZM ZW AM AZ BY KG MD RU TJ TM AT BE BG CH CY DE DK EE ES FI FR GB GR HU IE IS IT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW MR NE SN TD TG

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005810158

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2584906

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 182751

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 3081/DELNP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200580036519.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2007538330

Country of ref document: JP

Ref document number: 1020077009496

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005298840

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2005298840

Country of ref document: AU

Date of ref document: 20051026

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005298840

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2005810158

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11666171

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 11666171

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0518390

Country of ref document: BR