You are on page 1of 18

EMMI Intensive Programme "Design, Synthesis and Validation of Imaging Probes“

Torino, 19-30 September 2011

Basic principles and procedures


of solid phase peptide synthesis

Lorenzo Tei, PhD

Dipartimento di Scienze dell’Ambiente e della Vita


Università del Piemonte Orientale “Amedeo Avogadro”
Viale T. Michel 11, 15121, Alessandria

Contents:
 Introduction
 Historical background
 Different strategies for SPPS
 Possible problems
 Aminoacids and protections
 Amide bond formation
 Solid supports and linkers
 Analytical tests
 Peptide isolation and purification
WHY PEPTIDES?
Immune peptides: Hormones:
synthetic antigens; oxytocin Neuropeptides:
vaccines vasopressin cholecystokinin
diagnostic tools insulin neurotensin
immunostimulator peptides; somatostatin
muramyl dipeptide GnRH
etc. Antibiotics:
tuftsin derivatives
tachikinin
gramicidine S

Transporter peptides: Applications of


penetratin synthetic peptides Toxins:
oligoarginine conotoxins
HIV-Tat protein spider toxins
snake toxins
Carriers: ionchanel blockers
deliver diagnostic
Peptides
or therapeutic
for structural studies: Enzymes and
agents to
turn mimicking cyclic peptides enzyme inhibitors:
targeted cells
Ribonuclease A

Advantages of utilizing
peptides as therapeutic
molecules:

 High activity
 High specificity
 Unique 3D characteristics
 No accumulation in organs
 Low toxicity
 Less immunogenic than antibodies

 They can be made synthetically, by recombinant


methods or by chemical modification of an isolated
natural product.
Peptides developed for
therapeutic applications:
 Allergy/Asthma
 Arthritis
 Cancer
 Diabetes
 Cardiovascular diseases
 Inflammation
 Vaccines
 Ophtalmology
 Central Nervous System diseases
 Antiviral
 Antibacterial

Peptide synthesis:

Solution synthesis: Solid phase synthesis:


 Optimisation of reaction  Anchoring of the peptide
conditions and yields. chain on a resin.
 Purification of every step.  Use of excess reagents
 Time consuming. removed by filtration and
 Synthesis of small washing procedures.
peptides.  Simple, rapid and repetitive
 Useful for large-scale steps.
manufacturing.  High yields.
 Possible automation
 Synthesis of long peptides.
Beginnings of the SPPS

 Merrifield in 1963 introduced the use of a


polystyrenic support for the synthesis of peptides.
 Development of the Boc strategy (use of quite
strong acidic conditions)
 In the ’70 different polymeric supports and more
labile protecting groups were developed.
 In 1978 the Fmoc-based SPPS was introduced.
 In the last 15 years the Fmoc approach has
prevailed over the Merrifield SPPS.

Solid phase peptide synthesis

The idea: P P

anchoring
T AA1 X R T AA1 R

deprotection
P
P P P
T AA2
T AA2 AA1 R AA1 R
coupling (-H2O)

R = resin T = NH2 protecting P = side chain


group (boc, Fmoc) protecting group
P P

T AA2 AA1 R deprotection repetitive


cycle
coupling

P P P

T AAn AA2 AA1 R


cleavage
+
final deprotection

AAn AA2 AA1

Fmoc strategy in SPPS


R R
R' Polimer Attachment
Fmoc OH Fmoc O
NH
NH Polimer
O R' = OH, NH2, Cl O

Deprotection

O R R
NH O Coupling O
Fmoc NH Polimer H2N Polimer
R" O R" O
Fmoc OActivated
NH
Repeat deprotection
O
and coupling

O R
NH Cleavage
O
NH Polimer Peptide and Polimer
Fmoc n
R" O
Fmoc deprotection

O O
OH OH
O N O N
H H H H H
H O O N+
N

O
_ OH
N O N
H H
H H2+ O
N
N
+
OH
+ CO2 + H2N
O

Boc Strategy in SPPS


R
R
Boc OH Attachment
NH HO Polimer Boc O
NH Polimer
O
O
The Boc
Deprotection approach
needs to use
NH
O R
Coupling
R
highly toxic
O O
Boc NH Polimer H2N Polimer liquid HF for
R' O
R' O
Boc OActivated
cleavage
NH
Repeat deprotection
from the
O
and coupling resin.
O R
NH Cleavage
O Peptide and Polimer
Boc NH Polimer
n
R' O
Boc deprotection

H H
N O N O
R R
O O
H
H

R NH2 + CO2 H
N O +
R
+ H
O
H

Common AA side-chain
protecting groups used in
Fmoc SPPS
Side-chain fuctionality Commonly used Abbreviation
of amino acids protecting group

NH O
Arg H Pbf
N NH2 N
H S
O O

Asp, Glu, Ser, Thr, Tyr OH


tBu
O

Lys, Trp NH(2) N


(H)
O Boc

Asn, Gln, Cys, His XH


X trt
Coupling methods in
Fmoc SPPS
Pre-activation method:
R O
O
FmocHN N
 Pre-activation of the acid via O
activated esters. O
ONSu esters
 N-hydroxysuccinimide R F
(ONSu); slow formation of O F
FmocHN
the amide bond.
O
F F
 Pentafluorophenyl (OPfp) Pfp esters F
ester; in the presence of O
NHFmoc
HOBt the rate of reaction is
O
very rapid. R
N
HOBt esters N
N

Coupling methods in
Fmoc SPPS
In situ - activation method:
 Carbodiimides (DCC and DIC)
 Benzotriazole PF6 salts (HBTU, PyBOP, HATU)
(H3C)2N + N(CH3)2

N C N N
N PF6-
+N _
O
Dicyclohexylcarbodimide (DCC) HBTU

(H3C)2N + N(CH3)2
N
N
N P O N PF6- N PF6-
N N N
+N _ N
O
PyBOP HATU
Coupling using Diisopropylcarbodiimide

The use of HOBt minimises the formation of unreactive N-acylurea

Mechanism of amide
bond formation using
PyBOP/HOBt/DIPEA
(DIPEA)
OH N
H O-
Fmoc N H
O Fmoc N N
O
R N P O N PF6-
R N N
N

(PyBOP)
N N O-
N N
O
H N
Fmoc N N
O
( -OBt )
R

N O
N P O
NH2 R
N
NH2

Nu

NH
H
Fmoc N
O
R

+ O P N + -OBt
3
Solid supports:

 Dry polystyrene beads have an average diameter of


50 µm, but in DMF or DCM they swell 2.5 to 6.2 fold
in volume.
 SPPS takes place within a well solvated gel containing
mobile and reagent accessible peptide chains.
 Small particle sized resins with low cross linking
allows rapid diffusion of reagents inside the beads.
 Solid support and peptide backbone may be of
comparable polarities (polyamides, PEG).

Resins for SPPS

Resin Composition Functional group


Chloro/Aminomethyl 1% divinylbenzene cross-linked CH2Cl, CH2NH2,
polystyrene polystyrene benzhydrylamino

Tentagel Polystyrene-polyethylene glycol CH2OH, CH2NH2


graft polymer

O
Cl O n OH

Cl

O
Cl Cl O n OH

Merrifield Tentagel
Resins for SPPS

PEGA Bis 2-acrylamidoprop-1-ylPEG, 2-acrylamidoprop- CH2NH2


1-yl[2-aminoprop-1-yl]PEG dimethylacrylamide
co-polymer
NovagelTM (4’-O-methylpolyethylene glycoxycarbonylamino- CH2NH2
methyl)polystyrene

O
NMe2
O O
O HN O
HN O NH2
O n O n OCH3

HN O
O O
NH2
NH2
HN
O O
NH2

PEGA NovagelTM

Linkers:

The purpose of the linker are:


 provide a reversible linkage between the synthetic peptide
chain and the solid support.
 Protect the C-terminal α-carboxyl group during the process
of chain extension.
 Most linkers release peptide acids or amides upon
treatment with TFA.
 Some linkers permit the cleavage with nucleophiles, used
for the preparation of C-terminal modified peptides such as
esters secondary amides, aldehydes, alcohol.
Linkers:
Hydroxymethyl- Aminomethyl-based Trityl chloride-
based linkers linkers based linkers
OH
OMe NH2

O
Cl P

P Cl
MeO O P
Wang linker
(Hydroxymethylphenoxy) Rink linker (trialcoxybenzhydrylamine)

OMe 2-Chlorotrityl chloride


OMe
P O P O

OH
NH2 P
Sasrin linker OMe
NH
PAL linker (trialcoxybenzylamine) HO

H OH O
P N NH2

HMBA linker Bayer's 4-carboxytrityl linker


O O P
OCH3 OH

Sieber linker (9-Aminoxanthenyl)

H3CO O P

Rink acid linker

Resin handling procedures

 Apart from PEGA resin, all resins have to be


swollen before use.
 Underivatized polystyrene resins swell only
in dichloromethane.
 All the other are swollen in DMF.
 Resins (except PEGA) can be dried under
vacuum without damage.
 Resins are fragile and has to be handled
carefully (do not use magnetic stirrers!)
Attachment of 1st amino acid
For hydroxymethyl based linkers quite harsh
conditions has to be employed:
1. use of the symmetrical anhydride of the Fmoc
amino acid and DMAP in DMF.
2. Use of the Fmoc amino acid and dichlorobenzoyl
chloride in DMF/pyridine
HNFmoc O

R 0.1 eq. DMAP, DMF


1) O R O
R
OH
HNFmoc O
HNFmoc O
5 eq.

OH
R DCB (5 eq.), pyridine (8.25 eq.) O
2) R
HNFmoc O DCM OH
HNFmoc O
5 eq.

Attachment of 1st amino acid


Trityl based linkers:
OH
R 3 eq. DIPEA, DCM
1) R O
HNFmoc O
Cl HNFmoc O
1.2 eq
Cl

O Toluene, AcCl O
3 eq. DIPEA, DCM R
2) HN
OH
60°C 3h OH
R HNFmoc O

HNFmoc O
1.2 eq.

Aminomethyl-based linkers:
 Generally the amino function is Fmoc protected
 Standard piperidine deprotection and subsequent
coupling of the 1st amino acid
Synthesis protocol:

 Swelling of the resin


 1st Fmoc amino acid attachment
 Deprotection Fmoc group with piperidine in DMF
 Washings with DMF (between 5 to 10 times)
 Fmoc amino acid coupling (Activators/DIPEA/DMF)
 Washings with DMF
 Deprotection Fmoc and so on…
 After last coupling the resin has to be dried in
vacuo before cleavage

Analytical tests

Qualitative amine tests:


 Kaiser test (ninhydrin test)
http://www.natureprotocols.com/2007/10/24/monitoring_solid_phase_peptide.php

 TNBS test
 Chloranil test
http://www.chempep.com/ChemPep-Fmoc-Solid-Phase-Peptide-Synthesis.htm

Fmoc determination for estimation of first residue


attachment:
 The loading of the 1st AA can be calculated
determining the absorbance at 290 nm
(dibenzofulvene formed in the Fmoc deprotection)
Possible problems

Aggregation
Peptide chains on resin
can form secondary Enantiomerization
structures or aggregates During coupling the acidity of the H
either with other peptide atom on the α-carbon is enhanced
chains or with the by the carboxy group activation.
polymer support. Mainly May occur on attachment of Cys,
caused by H-bonding and His, Pro, Met, or Trp on
hydrophobic interactions hydroxymethyl-based resins
(high proportion of Ala,
Ile, Asn or Gln)

Side reactions
Rare if the AA are protected and the
correct procedures are followed.
Aspartimide formation can occur if Asp
is coupled to Gly, Ser, Asn, Thr or Arg.

Aspartimide formation
O
O Nu O
X Nu
O NH
NH N
N N NH O
H H N N
O O H H
O Nu

Hydrolysis

O O

OH NH
NH OH
N N
H H
O O

Base catalyzed aspartimide formation involves attack by the nitrogen atom


attached to the α-carboxy group of either aspartic acid or asparagine on the
side chain ester or amide group, respectively.
Nucleophilic attack then causes subsequent ring opening, which gives rise
to a mixture of α-aspartyl and β-aspartyl peptides.
Diketopiperazine formation

 Peptides containing proline or N-alkylated amino acids


attached via benzyl ester to the resin may have the
problem of diketopiperazine formation
 This not only causes a yield reduction but may also lead
to the generation of truncated sequences.
 In these cases is better to use a more hindered trityl resin
R R
O O
R'
N R'
H N
N N
O HO
O
O

Cleavage from the resin


 95% TFA for 1-3h cleaves the peptide from the
resin and deprotect t-butyl groups, Pbf group from
Arg, Trt group from Cys, Gln, Asn and His.
 Highly reactive cationic species are generated that
can react with amino acids containing electron rich
functional groups: Tyr, Trp, Met and Cys.
 Nucleophilic reagents (scavengers) are added to the
TFA to quench these ions: water, 1,2-Ethandithiol
(EDT), triisopropylsilane (TIS), thioanisole.
Cleavage cocktails
 If Cys is absent we normally use TFA, TIS 95:5 v/v.
 With Cys we add EDT (TFA, TIS, EDT; 95:3:2 v/v).
 With Met we add thioanisole (TFA, TIS, Thioanisole; 95:3:2 v/v)
 Reagent K (TFA, thioanisole, H2O, phenol, EDT; 82.5:5:5:5:2.5
v/v).
 Reagent R (TFA, thioanisole, anisole, EDT; 90:5:3:2).
 With Chlorotrityl chloride resins protections on the AA side-
chains can be mantained using DCM, AcOH, trifluoroethanol
80:10:10 v/v.
 With Trp a further step with a 5% water solution of AcOH is
required to obtain the fully deprotected peptide.

Peptide isolation and purification

 Precipitated by addition of cold diethyl ether


 Directly from TFA solution or following
evaporation of TFA and volatile scavengers.
 Centrifuge and wash with Et2O 3 times
 Dry the solid in vacuo
 Purification by HPLC
 Characterisation by MALDI-TOF mass
spectrum, NMR spectrometry.
Multisyntech SYRO II

Syro II automated peptide synthesizer

CEM Liberty

Liberty automated peptide synthesizer shown with single-mode


microwave reaction vessel

You might also like