BINDING INHIBITORS OF THE BETA. TRANSDUCIN REPEAT-CONTAINING PROTEIN

Abstract
The present invention relates to compounds which bind to Beta Trans-ducin repeat-containing protein (βTrCP), and modulate the activity of βTrCP. In particular, the invention relates to compounds which demonstrate optimised binding to PTrCP. The invention also relates to pharmaceutical compositions comprising such compounds and the use of such compounds as medicaments, specifically for the treatment of disorders associated with aberrant protein degradation, such as cancer. The preferred binding inhibitors are peptides derived from the motive DSGXXS, e.g. DEGFWE, DDGFWD and Succinyl-EGFWE.
Description
REFERENCE TO A “SEQUENCE LISTING” SUBMITTED AS ASCII TEXT FILES VIA EFS-WEB

The Sequence Listing written in file SEQ_098184-1092172.txt created on Sep. 11, 2018, 40,893 bytes, machine format IBM-PC, MS-Windows operating system, in accordance with 37 C.F.R. §§ 1.821 to 1.825, is hereby incorporated by reference in its entirety for all purposes.


FIELD OF THE INVENTION

The present invention relates to compounds which bind to Beta Transducin repeat-containing protein (βTrCP), and modulate the activity of βTrCP. In particular, the invention relates to compounds which demonstrate optimised binding to βTrCP. The invention also relates to pharmaceutical compositions comprising such compounds and the use of such compounds as medicaments, specifically for the treatment of disorders associated with aberrant protein degradation, such as cancer.


BACKGROUND OF THE INVENTION

In order to maintain the delicate homeostatic balance of a cell, unneeded or damaged proteins must be degraded. Protein degradation is performed by the proteasome, which dismantles unwanted proteins into small peptides of about eight amino acids in length. These peptides are then further degraded by proteases in the cell, and the resulting amino acids are used to synthesise new proteins.


To ensure that only unwanted proteins are degraded, and that healthy functioning proteins remain intact, target proteins are tagged for degradation by the ubiquitin proteasome system (UPS). The aim of the UPS is to attach a chain of approximately four ubiquitin monomers to any unwanted proteins in order to direct entry of the target protein into the proteasome.


The major components of the UPS are ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2) and ubiquitin ligases (E3). There are several members of each of these groups of enzymes, which generally recognise different groups of target proteins.


The first step of the UPS is the hydrolysation of ATP by an ubiquitin-activating enzyme in order to facilitate the adenylation of an ubiquitin molecule. Following this, the ubiquitin molecule is transferred to a cysteine residue in the active site of the ubiquitin-activating enzyme at the same time as a second ubiquitin molecule is adenylated. The second adenylated ubiquitin molecule is subsequently transferred to a cysteine residue in the active site of an ubiquitin-conjugating enzyme. The final step requires the recognition of the target protein by an ubiquitin ligase, which catalyses the transfer of the ubiquitin molecule from the ubiquitin-conjugating enzyme to the target protein. Following the addition of four ubiquitin molecules, the target protein is recognised by the proteasome, and sent for degradation.


βTrCP is an E3 ubiquitin ligase forming part of the UPS. It recognises a variety of target proteins, including inhibitor of nuclear factor κB (IκB), β-catenin, REST (repressor-element-1-silencing transcription factor), CDC25A/B, ATF4 (Activating Transcription Factor 4), and pro-caspase 3 and is known to function by binding to a phosphodegeneron motif DSGXXS in which the two serines are phosphorylated.


βTrCP is involved in apoptotic regulation through the targeted degradation of pro-apoptotic factors. βTrCP has been shown to be over-expressed in a variety of cancers including colorectal cancer, chemoresistant pancreatic cancer, hepatoblastomas and breast cancer. Human hepatocellular carcinomas (HCCs), pancreatic tumours and melanomas have also been shown to display an aberrant loss of IκB, which is thought to be caused by βTrCP over-expression. This over-expression increases the degradation of pro-apoptotic factors, leading to a reduction in apoptotic cell death and subsequent aberrant cell growth.


Inhibition of βTrCP prevents the degradation of pro-apoptotic factors such as IκB and programmed cell death 4 (PDCD4). This has been shown to induce apoptosis in human malignant melanoma, breast cancer and prostate cancer cells, augmenting the cytotoxic effects of anticancer drugs and ionizing radiation.


SUMMARY OF THE INVENTION

The inventors hypothesised that compounds that bond βTrCP may be able to prevent βTrCP binding to its substrates, thus preventing the ubiquitination of target proteins. The prolonged presence in a cell of pro-apoptotic factors will increase cellular apoptosis, providing a useful tool for the treatment of disorders associated with aberrant protein degradation such as hyperproliferative disorders including cancer.


The inventors have therefore designed a series of compounds which bind βTrCP and which will be therapeutically useful.


Compounds and Modified Peptides


The present invention relates to compounds which bind βTrCP.


Accordingly, in the first aspect, the invention provides a compound of Formula Ia:





X1-X2-X3-X4-X5-X6-X7   Formula Ia


wherein,


X1 is a group A1-B—Z1—;


X2 is a group —N(Ra)—Y1(-L1-A2)-Z2—;


X3 is a group —N(Rb)—Y2—Z3—;


X4 is a group —N(Rc)—Y3(-L2-A3)-Z4—; or X4 is a group —N(Rc)—Y3(-L2)-Z4


X5 is a group —N(Rd)—Y4(-L3-A4)-Z5—;


X6 is a group —N(Re)—Y5(-L4-A5)-Z6—;


X7 is a group —N(RN1)(RN2);


wherein,


B is C1-C10 alkyl, C2-C10 alkenyl, C2-C10 alkynyl or aryl;


wherein, B may be substituted with one or more RE, wherein RE is selected from the group consisting of C1-C4 alkyl, —NH2, —NH(RN2) and —N(RN2)2;


Ra, Rb, Rc, Rd, and Re are each independently selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


L1, L2, L3 and L4 are each independently C0-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl;


wherein,


L1 may be substituted with one or more RL1, wherein RL1 is C1-C4 alkyl;


L2 may be substituted with one or more RL2, wherein RL2 is C1-C4 alkyl or C2-C4 alkenyl;


L3 may be substituted with one or more RL3, wherein RL3 is C1-C4 alkyl;


L4 may be substituted with one or more RL4, wherein RL4 is C1-C4 alkyl;


Y1, Y3, Y4 and Y5 are each independently CH or N;


Y2 is CF2, CH2, N(RY2) or O; wherein, RY2 is —H or C1-C4 alkyl;


Z1 is a bond, C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) or C═NH;


Z2, Z3, Z4, Z5, and Z6 are independently selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH;


A1 and A5 are each independently carboxylic acid (—CO2H) or a bioisostere thereof and A2 is a carboxylic acid (—CO2H) or a bioisostere thereof or —C(O)N(RN1)2;


wherein,


A1 may be substituted with one or more RA1, wherein RA1 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl;


A2 may be substituted with one or more RA2, wherein RA2 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl;


A5 may be substituted with one or more RA5, wherein RA5 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl;


A3 and A4 are each independently aryl or heteroaryl; wherein, A3 may be substituted with one or more RA3, wherein, RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2;


A4 may be substituted with one or more RA4, wherein RA4 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl), —N(C1-C2 alkyl)2;


RN1 is selected from the group consisting of —H, C1-C10 alkyl and aryl;


RN2 is selected from the group consisting of RN1, —(CH2)0-10-(Z7)0-1-Aa, —(CH2O)0-10-CH2-(Z7)0-1-Aa, —(CH2CH2O)1-10-CH2CH3, —(CH2CH2O)1-10-(CH2)1-3-(Z7)0-1-Aa;


wherein,


Z7 is (C═O);


Aa is —OH, —NH2, —C(O)NH2, a cholesteryl derivative, a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids;


wherein,


when the compound of Formula Ia is substituted with an amino/amine group, said amino/amine group may be optionally capped, by replacement of a H atom, with a capping group.


Formula Ia may also be represented by Formula Ib:




embedded image


In a second aspect, the invention provides a modified peptide comprising a sequence of amino acids:





X1-E/D/pS-G-X4-X5-E/D/pS-NHRN2   Formula Ic


wherein,


each of the amino acids are selected from L-amino acids, D-amino acids, aza-amino acids and substituted amino acids; and


wherein,


X1 is a group A1-B—Z1—;


X4 is a group —N(Rc)—Y3(-L2-A3)-Z4—;


X5 is a group —N(Rd)—Y4(-L3-A4)-Z5—;


wherein,


B, Rc, Rd, L2, L3, Y3, Y4, Z1, Z4, Z5, A1, A3, A4, and RN2 are as previously defined.


In a third aspect, the invention provides a prodrug comprising a methyl, ethyl, propyl, butyl, pentyl, cyclopentyl, hexyl, benzyl, aryl or heteroaryl ester of a compound of Formula Ia or a modified peptide of Formula Ic.


In a fourth aspect, the invention provides a prodrug comprising a —CO2(CH2CH2O)1-10CH2CH3 ester of a compound of Formula Ia or a modified peptide of Formula Ic.


In a fifth aspect, the invention provides a pharmaceutical composition comprising a compound of Formula Ia or a modified peptide of Formula Ic; or a prodrug of a compound of Formula Ia or a modified peptide of Formula Ic.


In a sixth aspect, the invention provides a compound of Formula Ia, a modified peptide of Formula Ic, a prodrug of a compound of Formula Ia, a prodrug of a modified peptide of Formula Ic, or a pharmaceutical composition comprising a compound of Formula Ia or a modified peptide of Formula Ic, for use in medicine.


In a seventh aspect, the invention provides a compound of Formula Ia, a modified peptide of Formula Ic, a prodrug of a compound of Formula Ia, a prodrug of a modified peptide of Formula Ic, or a pharmaceutical composition comprising a compound of Formula Ia or a modified peptide of Formula Ic, for use in the treatment of a disease associated with aberrant protein degradation.


In an eighth aspect, the invention provides a method of treating a disease associated with aberrant protein degradation comprising administering a compound of Formula Ia, a modified peptide of Formula Ic, a prodrug of a compound of Formula Ia, a prodrug of a modified peptide of Formula Ic, or a pharmaceutical composition comprising a compound of Formula Ia or a modified peptide of Formula Ic, in a pharmaceutically effective amount.


In a ninth aspect, the invention provides a diagnostic kit comprising a compound of Formula Ia, a modified peptide of Formula Ic, a prodrug of a compound of Formula Ia, or a prodrug of a modified peptide of Formula Ic.


SUMMARY OF THE INVENTION

Embodiments of Compounds of Formula Ia


Various embodiments of the invention are described herein. It will be recognised that features specified in each embodiment may be combined with other specified features to provide further embodiments.


In the first aspect, the invention provides a compound of Formula Ia:





X1-X2-X3-X4-X5-X6-X7   Formula Ia


wherein, X1, X2, X3, X4, X5, X6 and X7 are as hereinbefore defined.


Carboxylic acid isosteres


Groups A1, A2 and A5 are each independently carboxylic acid (—CO2H) groups or bioisosteres thereof (and A2 can also be (˜C(O)N(RN1)2) “Bioisostere” is a term with which the skilled person will be familiar. In particular, bioisosteres (also known as non-classical isosteres) are functional groups or molecules which have chemical and physical similarities producing broadly similar biological properties to those of the replaced moiety (Stocks et al. On Medicinal Chemistry, 2007).


Carboxylic acids are weak organic acids with pKas in the range of 0-5, although this can be affected by the electronegative or electropositive nature of any substituents. For example, acetic acid (CH3CO2H) has a pKa of 4.8. Bioisosteres of carboxylic acids may have comparable pKa values to those of carboxylic acids, i.e. they may be deprotonated at physiological pH (pH 7.3-7.5, Werle et al. British Journal of Cancer, 1997).


Common bioisosteric replacements for carboxylic acids include functional groups such as sulfonamides (pKa ˜4-9), sulfamides (pKa ˜6-10), acylsulfonamides (pKa ˜5), sulfonyl ureas (pKa ˜3-5), hydroxaminc acids (pKa ˜9), acylcyanamides (pKa ˜8), sulfonic acids (pKa ˜2), sulfonates (pka ˜1-2), phosphates (pKa ˜2), phosphonic acids/phosphonates (pKa ˜6.5) and phosphinic acids (pKa ˜4). Heterocycles with intrinsic acidity may also be used as bioisosteres for carboxylic acids. Common heterocyclic bioisosteric replacements for carboxylic acids include tetrazoles (pKa ˜4-8), triazoles (pKa ˜9), isoxazolones (pKa ˜5), 1,2,4-oxadiazolones (pKa ˜6), and 1,2-dihydro-pyrazolones (pKa ˜8). Examples of carboxylic acid bioisosteric functional groups include:




embedded image


wherein, R1 is RA1, RA2 or RA5 respectively, wherein RA1, RA2 and RA5 are as previously defined.


Examples of heterocyclic carboxylic acid bioisosteres include:




embedded image


wherein, R1 is RA1, RA2 or RA5 respectively, wherein RA1, RA2 and RA5 are as previously defined.


Group X1


It is believed that the side chain of group X1 interacts with the βTrCP binding domain to form an ionic bridge. Accordingly, X1 is a functional group which is ionisable at physiological pH, in particular a carboxylic acid group or bioisostere thereof, in order to sustain such a binding interaction with the βTrCP binding domain.


X1 is a group A1-B—Z1—;


wherein,


A1 is carboxylic acid (˜CO2H) or a bioisostere thereof;


wherein, A1 may be substituted with one or more RA1, wherein RA1 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl;


B is C1-C10 alkyl, C2-C10 alkenyl or C2-C10 alkynyl or aryl;


wherein, B may be substituted with one or more RE, wherein RE is selected from the group consisting of C1-C4 alkyl, —NH2, —NH(RN2) and —N(RN2)2; and


Z1 is a bond, C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) or C═NH.


The term bioisostere is as hereinbefore described. In one embodiment, A1 is carboxylic acid. In one embodiment, A1 is selected from the group consisting of




embedded image


embedded image


embedded image


wherein, R1 is RA1.


In one embodiment, A1 is selected from the group consisting of




embedded image


wherein, R1 is RA1.


In one embodiment, A1 is selected from the group consisting of




embedded image


embedded image


wherein, R1 is RA1.


In one embodiment, A1 is selected from the group consisting of carboxylic acid (—CO2H), phosphate, phosphonate, phosphonic acid, tetrazole and sulphate.


In one embodiment, A1 is selected from the group consisting of carboxylic acid and phosphate. In one embodiment, preferably A1 is carboxylic acid.


In one embodiment, A1 is substituted by RA1, wherein RA1 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl.


In one embodiment, B is C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl. In one embodiment, B is C1-C2 alkyl or C2 alkenyl. In one embodiment B is aryl, particularly B is phenyl.


In one embodiment, B is substituted with one or more RE, wherein RE is selected from the group consisting of C1-C4 alkyl, —NH2, —NH(RN2) and —N(RN2)2. In one embodiment, B is substituted with —NH2. In one embodiment, B is substituted with —NH(RN2), wherein RN2 is a chain of one or more naturally or non-naturally occurring amino acids.


In one embodiment, B is substituted with —N(RN2)2. In one embodiment, B is substituted with —N(RN2)2, wherein both RN2 are RN1, wherein one RN1 is —H and the other RN1 is C1-C10 alkyl, aryl or heteroaryl.


In one embodiment, Z1 is C═O, C═S, or CH2. In one embodiment, Z1 is C═O.


In one embodiment, X1 is of Formula HO2C—B—Z1—; wherein Z1 is a bond, C═O, C═S, CH2, S═O or S(O)2; and B is C1-C10 alkyl, C2-C10 alkenyl or C2-C10 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z—, wherein Z1 is C═O and B is C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C1 alkyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkyl, C2 alkenyl or C2 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C3 alkyl, C3 alkenyl or C3 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C4 alkyl, C4 alkenyl or C4 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C5 alkyl, C5 alkenyl or C5 alkynyl. In one embodiment, X1 is of Formula HO2C—B—Z—, wherein Z1 is C═O and B is C6 alkyl, C6 alkenyl or C6 alkynyl. In one embodiment, X1 is of Formula HO2C-E-Z1—, wherein Z1 is C═O and B is C2-alkyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkenyl. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkyl; wherein C2 alkyl is substituted with one or more RE. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkyl; wherein C2 alkyl is substituted with one or more —N(RN2)2. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkenyl; wherein C2 alkenyl is substituted with one or more —N(RN2)2. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C3 alkyl; wherein C3 alkyl is substituted with one or more —N(RN2)2. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C3 alkyl; wherein C3 alkyl is substituted with one or more —N(RN1)(RN2). In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C3 alkenyl; wherein C3 alkenyl is substituted with one or more —N(RN1)(RN2). In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C3 alkyl; wherein C3 alkyl is substituted with —NH2. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 alkyl; wherein C2 alkyl is substituted with —NH2 at the carbon atom adjacent to Z1. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 or C3 alkyl, wherein said C2 or C3 alkyl is substituted with one or more —N(RN1)(RN2) wherein RN2 is a chain of one or more amino acids. In one embodiment, X1 is of Formula HO2C—B—Z1—, wherein Z1 is C═O and B is C2 or C3 alkyl, wherein said C2 or C3 alkyl is substituted with one or more —N(RN1)(RN2) wherein RN1 is —H and RN2 is a chain of one or more naturally or non-naturally occurring amino acids.


In another embodiment, X1 may be selected from the group consisting of:




embedded image


In one embodiment, X1 is aspartyl, succinyl or maleyl. In one embodiment, preferably X1 is aspartyl. In one embodiment, X1 is aspartyl or glutamyl, and is substituted at the N-terminus with a chain of one or more naturally or non-naturally occurring amino acids.


When X1 is aspartyl, it is preferably L-aspartyl (D) or D-aspartyl (d). When X1 is glutamyl it is preferably L-glutamyl (E) or D-glutamyl (e).


Group X2


It is believed that the side chain of group X2 interacts with the βTrCP binding domain to form an ionic bridge. Accordingly, X2 is a functional group which is ionisable at physiological pH, in particular carboxylic acid groups or bioisosteres thereof, in order to sustain such a binding interaction with the βTrCP binding domain.


X2 is a group —N(Ra) —Y1(-L1-A2)-Z2—;


wherein;


Ra is selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


L1 is C0-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl; wherein, L1 may be substituted with one or more RL1, wherein RL1 is C1-C4 alkyl;


Y1 is CH or N;


Z2 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH; and


A2 is carboxylic acid (—CO2H) or a bioisostere thereof or —C(O)N(RN1)2; wherein, A2 may be substituted with one or more RA2, wherein RA2 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl.


In one embodiment, Ra is —H. In one embodiment, Ra is C1-C10 alkyl.


In one embodiment, Y1 is CH. In one embodiment, Y1 is N.


In one embodiment, A2 is carboxylic acid. In one embodiment, A2 is selected from the group consisting of




embedded image


wherein, R1 is RA2.


In one embodiment, A2 is selected from the group consisting of




embedded image


wherein, R1 is RA2.


In one embodiment, A2 is selected from the group consisting of




embedded image


embedded image


wherein, R1 is RA2.


In one embodiment, A2 is selected from the group consisting of carboxylic acid (—CO2H), phosphate, phosphonate, phosphonic acid, tetrazole and sulphate.


In one embodiment, A2 is selected from the group consisting of carboxylic acid and phosphate. In one embodiment, preferably A2 is carboxylic acid.


In one embodiment, A2 is substituted with one or more RA2, wherein RA2 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl. In one embodiment, A2 is substituted with one or more RA2, wherein RA2 is methyl or ethyl. In one embodiment, A2 is substituted with one or more RA2, wherein RA2 is methyl.


In one embodiment A2 is —C(O)N(RN1)2 wherein each RN1 may be the same or different. Particularly A2 is C(O)NH(RN1), more particularly A2 is C(O)NH2


In one embodiment, L1 is C0-C5 alkyl or C2-C5 alkenyl. In one embodiment, L1 is C0-C5 alkyl. In one embodiment, L1 is preferably C1-C2 alkyl.


In one embodiment, L1 is substituted with one or more RL1, wherein RL1 is C1-C4 alkyl. In one embodiment, L1 is substituted with one or more RL1, wherein RL1 is methyl.


In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1-C5 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1-C2 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C2 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H). In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1 alkyl, Y1 is CH, Z2 is C═O and A2 is phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C2 alkyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H). In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C2 alkyl, Y1 is CH, Z2 is C═O and A2 is phosphate. In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1 alkyl substituted with RL1, wherein RL1 is methyl, Y1 is CH, Z2 is C═O and A2 is carboxylic acid (—CO2H). In one embodiment, X2 is of Formula —NH—Y1(-L1-A2)-Z2—; wherein L1 is C1 alkyl substituted with RL1, wherein RL1 is methyl, Y1 is CH, Z2 is C═O and A2 is phosphate.


In one embodiment, group X2 may be a glutamate, an aspartate, or a phosphorylated serine residue. In one embodiment, preferably, X2 is glutamate or aspartate. In one embodiment, the glutamate, aspartate, or phosphorylated serine residue of group X2 is an L-amino acid. In a further embodiment, X2 is phosphorylated threonine.


In one embodiment, preferably, group X2 is a glutamate or aspartate residue. This eliminates the requirement for phosphorylated serine residues, which are naturally present within the phosphodegeneron sequence, whilst retaining binding. The negatively charged phosphorylated serine residues are not synthetically desirable.


In one embodiment, the glutamate, aspartate or phosphorylated serine residue of X2 may be substituted with methyl. In another embodiment, the glutamate, aspartate or phosphorylated serine residue of group X2 may be substituted with ethyl.


Group X3


It is believed that group X3 associates with an area in the βTrCP binding domain which may accommodate a compound/modified peptide with a beta-turn.


Accordingly, X3 is a functional group which is suitably configured to reside in this area of the βTrCP binding domain.


X3 is a group —N(Rb)—Y2—Z3—;


wherein,


Rb is selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


Y2 is CF2, CH2, N(RY2) or O; wherein, RY2 is —H or C1-C4 alkyl; and


Z3 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH.


In one embodiment, Rb is —H or C1-C10 alkyl. In one embodiment, preferably Rb is —H.


In one embodiment, Y2 is CH2 or N(RY2). In one embodiment, Y2 is preferably CH2.


In one embodiment, Y2 is N(RY2). In one embodiment, Y2 is N(RY2), wherein RY2 is methyl.


In one embodiment, X3 is of Formula —NH—Y2—Z3—, wherein Y2 is CH2, N(RY2) or O and Z3 is selected from the group consisting of C═O, C═S, CH2, S═O and S(O)2. In one embodiment, preferably, X3 is of Formula —NH—Y2—Z3—, wherein Y2 is CH2 and Z3 is C═O. In one embodiment, X3 is of Formula —NH—Y2—Z3—, wherein Y2 is NH and Z3 is C═O.


In one embodiment, preferably group X3 is a glycine residue.


In one embodiment the glycine residue of group X3 is an aza glycine residue, wherein an “aza amino acid” is an L-/D-amino acid in which the α-carbon atom has been replaced by a nitrogen atom.


In one embodiment the glycine residue of group X3 is an oxo glycine residue, wherein an “oxo amino acid” is an L-/D-amino acid in which the α-carbon atom has been replaced by an oxygen atom.


Group X4


Without wishing to be bound by theory, it is believed that the side chain of group X4 sustains a Van der Waals interaction with the βTrCP binding domain.


X4 is a group —N(Rc)—Y3(-L2-A3)-Z4—; or —N(Rc)—Y3(-L2)-Z4


wherein,


Rc is selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


L2 is C0-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl; wherein, L2 may be substituted with one or more RL2, wherein RL2 is C1-C4 alkyl or C2-C4 alkenyl;


Y3 is CH or N;


Z4 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH; and


A3 is aryl or heteroaryl; wherein, A3 may be substituted with one or more RA3, wherein, RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2;


In one embodiment, X4 is a group —N(Rc)—Y3(-L2-A3)-Z4—.


In one embodiment, X4 is a group —N(Rc)—Y3(-L2)-Z4—.


In one embodiment, Rc is —H or C1-C10 alkyl. In one embodiment, preferably Rc is —H.


In one embodiment, Y3 is CH. In one embodiment, Y3 is N.


In one embodiment, L2 is C0-C5 alkyl or C2-C5 alkenyl. In one embodiment, L2 is C0-C5 alkyl. In one embodiment, L2 is C1-C2 alkyl. In one embodiment, preferably L2 is C1 alkyl.


In one embodiment, L2 is substituted with one or more RL2, wherein RL2 is C1-C4 alkyl or C2-C4 alkenyl. In one embodiment, L2 is substituted with RL2, wherein RL2 is methyl.


In one embodiment, A3 is aryl. In one embodiment, A3 is phenyl. In one embodiment, A3 is heteroaryl. In one embodiment, A3 is aryl or heteroaryl substituted with one or more RA3, wherein, RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2.


In one embodiment, A3 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl.


In one embodiment, A3 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —F, —Cl, —OH and —NO2.


In one embodiment, A3 is phenyl substituted at one or more of the 2-, 3- or 4-positions, with RA3, wherein RA3 is a substituent selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H and —C1-C10 alkyl.


In one embodiment, preferably A3 is phenyl substituted with one or more RA3, wherein RA3 is selected from the group consisting of —F, —Cl, —NO2 and —OH.


In another embodiment, preferably A3 is phenyl substituted with RA3, wherein RA3 is chlorine and/or fluorine.


In one embodiment, Z4 is selected from the group consisting of C═O, C═S and CH2.


In one embodiment, Z4 is C═O. In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1-C5 alkyl and A3 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with one or more RA3.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1-C5 alkyl and A3 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1-C5 alkyl and A3 is aryl or heteroaryl, wherein said aryl or heteroaryl is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), C1-C10 alkyl and —NO2.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1 alkyl and A3 is phenyl, wherein said phenyl is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl) and —NO2.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1 alkyl and A3 is selected from the group consisting of indole and imidazole, wherein said indole or imidazole is substituted with one or more RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), C1-C10 alkyl and —NO2.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1 alkyl and A3 is phenyl, wherein said phenyl is substituted at one or more of the 2-, 3- or 4-positions with RA3, wherein RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), C1-C10 alkyl and —NO2.


In one embodiment, X4 is of Formula —NH—Y3(-L2-A3)-Z4—; wherein Y3 is CH, Z4 is C═O, L2 is C1 alkyl and A3 is phenyl, wherein said phenyl is substituted at one or more of the 2-, 3- or 4-positions with RA3, wherein RA3 is selected from the group consisting of —F, —Cl, —NO2 and —OH.


In one embodiment, group X4 is an aromatic alanine derivative.


In one embodiment, preferably group X4 is selected from the group consisting of phenylalanine, tyrosine, tryptophan and histidine. In one embodiment, X4 is phenylalanine. In one embodiment, group X4 may be an L amino acid. In one embodiment, group X4 is selected from the group consisting of phenylalanine, tyrosine, tryptophan and histidine, wherein said phenylalanine, tyrosine, tryptophan and histidine is substituted with one or more RA3.


In one embodiment, group X4 is selected from the group consisting of:




embedded image


Group X5


It is believed that the side chain of group X5 interacts with an alkyl portion within the βTrCP binding domain. Accordingly, X5 is a functional group which is capable of sustaining such an interaction within the βTrCP binding domain.


X5 is a group —N(Rd)—Y4(-L3-A4)-Z5—;


wherein;


Rd is selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


Y4 is CH or N;


L3 is C0-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl; wherein, L3 may be substituted with one or more RL3, wherein RL3 is C1-C4 alkyl;


A4 is aryl or heteroaryl; wherein, A4 may be substituted with one or more RA4, wherein RA4 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2; and


Z5 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH.


In one embodiment, Rd is —H or C1-C10 alkyl. In one embodiment, preferably Rd is —H.


In one embodiment, Y4 is CH. In one embodiment, Y4 is N.


In one embodiment, L3 is C0-C5 alkyl or C2-C5 alkenyl. In one embodiment, L3 is C0-C5 alkyl. In one embodiment, L3 is C1-C2 alkyl. In one embodiment, preferably L3 is C1 alkyl.


In one embodiment, L3 is substituted with RL3, wherein RL3 is C1-C4 alkyl. In one embodiment, L3 is substituted with RL3, wherein RL3 is methyl.


A4 is aryl or heteroaryl. In one embodiment, A4 is aryl. In one embodiment, A4 is bi-aryl, monocyclic aryl or polycyclic fused ring aryl. In one embodiment, A4 is heteroaryl. In one embodiment, A4 is monocyclic heteroaryl. In one embodiment, A4 is polycyclic fused ring heteroaryl.


In one embodiment, A4 is selected from the group consisting of:




embedded image


In one embodiment, A4 is selected from the group consisting of phenyl, biphenyl, naphthyl, indenyl, fluorenyl, anthracyl and phenanthryl. In one embodiment, A4 is selected from the group consisting of pyridyl, thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazoyl, oxadiazolyl, thiadiazolyl and tetrazolyl. In one embodiment, A4 is selected from the group consisting of indolyl, benzofuranyl, quinolyl, isoquinolyl, indazolyl, indolinyl, isoindolyl, indolizinyl, benzimidazolyl or quinolinyl. In one embodiment, A4 is selected from the group consisting of phenyl, naphthyl, indolyl and imidazoyl.


In one embodiment, A4 is aryl or heteroaryl, wherein said aryl or heteroaryl are substituted with one or more RA4, wherein RA4 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —C1-C10 alkyl, —NH2, —NH(C1-C2 alkyl) and —N(C1-C2 alkyl)2.


In one embodiment, X5 is of Formula —NH—Y4(-L3-A4)-Z5—; wherein Y4 is CH, Z5 is C═O, L3 is C1-C5 alkyl and A4 is aryl. In one embodiment, X5 is of Formula —NH—Y4(-L3-A4)-Z5—; wherein Y4 is CH, Z5 is C═O, L3 is C1-C5 alkyl and A4 is heteroaryl.


In one embodiment, preferably, X5 is of Formula —NH—Y4(-L3-A4)-Z5—; wherein Y4 is CH, Z5 is C═O, L3 is C1 alkyl and A4 is selected from the group consisting of phenyl, naphthyl, indolyl and imidazoyl.


In one embodiment, preferably group X5 is selected from the group consisting of tryptophan, naphthyl-alanine, histidine and phenylalanine, wherein X5 may be substituted at one or more positions with RA4. In one embodiment, group X5 is selected from the group consisting of tryptophan, 1 naphthyl-alanine, 2 napthyl-alanine, histidine and F(4NO2). In one embodiment, group X5 is tryptophan. In one embodiment, group X5 is tryptophan, wherein the nitrogen of the indole group is substituted with methyl. In one embodiment, group X5 is naphthyl-alanine. In one embodiment, group X5 is 2 naphthyl-alanine. In one embodiment, group X5 is 1 naphthyl-alanine. In one embodiment, group X5 is histidine. In one embodiment, group X5 is phenylalanine. In one embodiment, group X5 is phenylalanine, wherein in phenyl is substituted with one or more RA4. In one embodiment, group X5 is F(4NO2) or F(3NO2). In one embodiment, group X5 is an L amino acid. In one embodiment, the tryptophan, naphthyl-alanine, histidine or phenylalanine residue of group X5 may be substituted at one or more positions with RA4.


Group X6


It is believed that the side chain of group X6 interacts with the βTrCP binding domain to form an ionic bridge. Accordingly, X6 is a functional group which is ionisable at physiological pH, in particular a carboxylic acid group or bioisostere thereof, in order to sustain such a binding interaction with the βTrCP binding domain.


X6 is a group —N(Re)—Y5(-L4-A5)-Z6—;


wherein;


Re is selected from the group consisting of —H, C1-C10 alkyl, aryl and heteroaryl;


L4 is C0-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl; wherein L4 may be substituted with one or more RL4, wherein RL4 is C1-C4 alkyl;


Y5 is CH or N;


Z6 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4 alkyl) and C═NH; and


A5 is carboxylic acid (—CO2H) or a bioisostere thereof; wherein, A5 may be substituted with one or more RA5, wherein RA5 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl.


In one embodiment, Re is —H. In one embodiment, Re is C1-C10 alkyl. In one embodiment, Re is C1-C4 alkyl. In one embodiment, Re is methyl or ethyl. In one embodiment, Re is methyl. In one embodiment, Re is aryl or heteroaryl. In one embodiment, Re is phenyl.


In one embodiment, Y5 is CH. In one embodiment, Y5 is N.


In one embodiment, L4 is C0-C5 alkyl or C2-C5 alkenyl. In one embodiment, L4 is C0-C5 alkyl. In one embodiment, L4 is preferably C1-C2 alkyl.


In one embodiment, L4 is substituted with one or more RL4, wherein RL4 is C1-C4 alkyl.


In one embodiment, A5 is carboxylic acid. In one embodiment, A5 is selected from the group consisting of




embedded image


embedded image


embedded image


wherein, R1 is RA5.


In one embodiment, A5 is selected from the group consisting of




embedded image


wherein, R1 is RA5.


In one embodiment, A5 is selected from the group consisting of




embedded image


embedded image


wherein, R1 is RA5.


In one embodiment, A5 is selected from the group consisting of carboxylic acid (—CO2H), phosphate, phosphonate, phosphonic acid, tetrazole and sulphate.


In one embodiment, A5 is selected from the group consisting of carboxylic acid and phosphate. In one embodiment, preferably, A5 is carboxylic acid.


In one embodiment, A5 is substituted with one or more RA5, wherein RA5 is selected from the group consisting of —H, C1-C4 alkyl, C2-C4 alkenyl and aryl.


In one embodiment, X6 is of Formula —N(Re)—Y5(-L4-A5)-Z6—; wherein Re is —H, C1-C10 alkyl, aryl or heteroaryl, L4 is C1-C5 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —N(Re)—Y5(-L4-A5)-Z6—; wherein Re is —H, C1-C10 alkyl, aryl or heteroaryl, L4 is C1-C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —N(Re)—Y5(-L4-A5)-Z6—; wherein Re is —H, C1-C4 alkyl or aryl, L4 is C1-C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —N(Re)—Y5(-L4-A5)-Z6—; wherein Re is methyl, L4 is C1-C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H). In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is phosphate. In one embodiment, X6 is of Formula —N(CH3)—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —N(CH3)—Y5(-L4-A5)-Z6—; wherein L4 is C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H) or phosphate. In one embodiment, X6 is of Formula —N(CH3)—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H). In one embodiment, X6 is of Formula —N(CH3)—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl, Y5 is CH, Z6 is C═O and A5 is phosphate. In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C2 alkyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H). In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C2 alkyl, Y5 is CH, Z6 is C═O and A5 is phosphate. In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl substituted by methyl, Y5 is CH, Z6 is C═O and A5 is carboxylic acid (—CO2H). In one embodiment, X6 is of Formula —NH—Y5(-L4-A5)-Z6—; wherein L4 is C1 alkyl substituted by methyl, Y5 is CH, Z6 is C═O and A5 is phosphate.


In one embodiment, group X6 may be a glutamate, an aspartate or a phosphorylated serine residue. In one embodiment, the glutamate, aspartate or phorphorylated serine residue of group X6 is an L amino acid. In a further embodiment, X6 may be phosphorylated threonine.


In one embodiment, preferably group X6 is a glutamate or aspartate residue. This eliminates the requirement for phosphorylated serine residues, which are naturally present within the phosphodegeneron sequence, whilst retaining binding. The negatively charged phosphorylated serine residues are not synthetically desirable.


In one embodiment, the glutamate, aspartate or phosphorylated serine residue of X6 may be substituted with methyl. In another embodiment, the glutamate, aspartate or phosphorylated serine residue of group X6 may be substituted with ethyl.


Group X7


It is believed that group X7 forms a hydrogen bond with the βTrCP binding domain. Accordingly, X7 is a functional group which is capable of forming such a hydrogen bond with the βTrCP binding domain.


X7 is a group —N(RN1)(RN2); wherein RN1 and RN2 are as previously defined. In one embodiment, preferably X7 is —NH2. In one embodiment, X7 is —N(RN1)2, wherein one RN1 is —H and the other RN1 is C1-C10 alkyl or aryl. In one embodiment, X7 is —N(RN1)2, wherein both RN1 are independently C1-C10 alkyl or aryl.


In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z)0-1-Aa, and wherein Aa is —OH. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z7)0-1-Aa, and wherein Aa is —NH2. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)4-8-(Z7)0-1-Aa, and wherein Aa is —NH2. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z7)0-1-Aa, and wherein Aa is —C(O)NH2. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z7)0-1-Aa, and wherein Aa is a chain of one or more naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z7)0-1-Aa, and wherein Aa is a chain of one or more non-naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10-(Z)0-1-Aa, and wherein Aa is a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2)0-10—(Z7)0-1-Aa, and wherein Aa is a cholesteryl derivative.


In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2CH2O)1-10-CH2CH3. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2CH2O)1-10-(CH2)1-3-(Z7)0-1-Aa, and wherein Aa is —NH2 or —C(O)NH2. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2 is —(CH2CH2O)1-10-(CH2)1-3-(Z7)0-1-Aa, and wherein Aa is a chain of one or more naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2-(CH2CH2O)4-8-(CH2)1-3-(Z7)0-1-Aa, and wherein Aa is a chain of one or more naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2-(CH2CH2O)1-10-(CH2)1-3-(Z7)0-1-Aa, and wherein Aa is a chain of one or more non-naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2-(CH2CH2O)4-8-(CH2)1-3-(Z7)0-1-Aa, and wherein Aa is a chain of one or more non-naturally occurring amino acids. In one embodiment, X7 is —N(RN1)(RN2), wherein RN2-(CH2CH2O)1-10-(CH2)1-3-(Z7)0-10-Aa, and wherein Aa is a cholesteryl derivative. In one embodiment RN2 is —(CH2)0-10-(Z)0-1-Aa and Aa is a cholesteryl derivative, in particular the cholesteryl derivative is:




embedded image


wherein RN1 is as previously defined, Y6 is CH or N and Z8 is selected from the group consisting of C═O, C═S, CH2, S═O, S(O)2, C═N(C1-C4) alkyl) and C═NH. In particular, the cholesteryl derivative is:




embedded image


It is believed that the cholesteryl group enhances the cell penetration of the compounds and modified peptides of the invention, without affecting the activity of the compounds and modified peptides of the invention against the targets of the invention.


Additional Amino Acids/Chains of Substituents


Compounds of the invention may additionally contain one or two chains of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids. For example, in one embodiment, group B may be substituted with —NH(RN2) or —N(RN2)2, wherein one RN2 is a chain of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more naturally occurring or non-naturally occurring amino acids. Alternatively, group X7 may be of Formula —N(RN1)(RN2), wherein RN1 is preferably —H and RN2 is a chain of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more naturally or non-naturally occurring amino acids.


Alternatively, group E may be substituted with —NH(RN2) or —N(RN2)2, wherein one RN2 is a chain of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more naturally occurring or non-naturally occurring amino acids and X7 may be of Formula —N(RN1)(RN2), wherein RN2 is a chain of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids, thus providing a compound containing two additional chains of amino acids. The one or more naturally occurring or non-naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids.


Chains of non-naturally occurring amino acids may include peptoids, which are peptidomimetics whose side chains are appended to the nitrogen atom of the peptide, rather than to the alpha-carbons.


Modified Peptide Embodiment of Formula Ia


In one embodiment, the compound of Formula Ia may comprise a sequence of amino acids according to the following Formula Ic:





X1-E/D/pS-G-X4-X5-E/D/pS-NHRN2   Formula Ic


X1, X4, X5 and RN2 are as previously defined.


In one embodiment, the compound of Formula Ia may comprise a sequence of amino acids according to the following Formula Iv:






d-E-G-F(3F)—W-E-NHRN2   Formula Iv


Herein “comprise” is used in the open sense to indicate that additional amino acids may also be present in the sequence.


The amino acids of the Formula depicted above preferably form a contiguous sequence.


In order to arrive at Formula Ic, the inventors used the phosphodegeneron sequence as a starting point, and systematically substituted each of the amino acids to alternative natural and non-natural amino acids. At each stage the binding of the substituted peptides to βTrCP was assessed and further substituted peptides were designed in order to maximise binding.


Within the meaning of the present invention, the term “modified” indicates that the peptide is not naturally occurring. A modified peptide may contain one or more non-naturally occurring amino acids, and/or may include one or more moieties which are not classified as amino acids.


Within the present invention, the term “residue” will be used to refer to each of the component moieties of the modified peptide, whether these are amino acids or other chemical moieties. Within Formula Ic, the individual residues are shown separated by hyphens (“-”).


Where the residues of the modified peptide are amino acids, each of the amino acids may be independently selected from an L-amino acid, a D-amino acid and an aza-amino acid. One or more of the residues may additionally be independently substituted at one or more positions irrespective of which subtype of amino acid forms the basis for the residue.


An “L amino acid” is defined as an amino acid which can theoretically be synthesised from levorotatory glyceraldehyde. Amino acids found in naturally occurring proteins are usually L amino acids. According to generally accepted notation, L amino acids are depicted herein using the capital letter single letter amino acid code.


A “D amino acid” is the stereoisomer of an L amino acid and is defined as an amino acid which can theoretically be synthesised from dextrorotary glyceraldehyde.


According to generally accepted notation, D amino acids are depicted herein using the lower case single letter amino acid code.


An “aza amino acid” is an L amino acid in which the α-carbon atom has been replaced by a nitrogen atom. The replacement of the αC—COOH bond found in naturally occurring amino acids with an αN—COOH bond can increase the stability of a peptide.


Herein, the suffix “p” is used to denote a phosphorylated residue, e.g. “pS” denotes phosphorylated serine.


The compounds of the present invention bind to βTrCP. In one embodiment the compounds are considered to “bind to βTrCP” if they bind with an affinity of less than about 10M. In some embodiments the compounds/peptides may bind to βTrCP with an affinity of less than about 900 nM, less than about 800 nM, less than about 700 nM, less than about 600 nM, less than about 500 nM, less than about 400 nM, less than about 300 nM, less than about 200 nM, less than about 150 nM, less than about 100 nM, less than about 90 nM, less than about 80 nM, less than about 70 nM, less than about 60 nM, less than about 50 nM, less than about 40 nM, less than about 30 nM, less than about 20 nM, less than about 10 nM, less than about 9 nM, less than about 8 nM, less than about 7 nM, less than about 6 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, less than about 1 nM or less.


Capping Groups


The compounds or modified peptides of the present invention may comprise a capping group. The function of the capping group is to increase the stability of the compound towards enzymic degradation, thus improving cell penetration, and any groups which are known to perform this function may be used as capping groups. In embodiments where a capping group is present, the definitions given for compounds of Formula Ia, Ib and Ic above equally apply.


In one embodiment, any amino/amine group, in particular an —NH2, —NH(RN1), or —NH(RN2) group which is present in a compound of the present invention may be capped, by replacement of a H atom with a capping group. Suitable capping groups include any groups which are known to prevent the compound from being degraded on entry into a cell.


In one embodiment, the capping group may be selected from the group consisting of




embedded image


and the thiocarbonyl derivatives of any of these capping groups.


The person skilled in the art will recognise that R* is used to indicate a generic structure for the purposes of illustrating the various functional groups which may be suitable as amine/amino capping groups. Specific examples of capping groups are illustrated below.


In one embodiment, a compound of the present invention is substituted by an amino/amine group, in particular an —NH2, —NH(RN1), or —NH(RN2) group as defined previously, wherein said amino/amine group, in particular the —NH2, —NH(RN1), or —NH(RN2) group, is capped, by the replacement of a H atom with a capping group selected from the group consisting of:




embedded image


wherein,


Rcg is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —CN, —NO2, —CF3, —OCF3, —CO2H, —NH2, —NH(C1-C2 alkyl), —N(C1-C2 alkyl)2, —C1-C10 alkyl, aryl and heteroaryl.


In a further embodiment, the capping group may be selected from the group consisting of:




embedded image


embedded image


embedded image


Further capping groups which may be used in the synthesis of compounds of the present invention are:




embedded image


embedded image


Acidic capping groups which may be used in the synthesis of compounds of the present invention are:




embedded image


embedded image


In one embodiment, group B is substituted by a substituent of Formula —NH2, —NH(RN1), or —NH(RN2), wherein the —NH2, —NH(RN1) or —NH(RN2) substituent is capped, by the replacement of a H atom, with a capping group selected from the group consisting of:




embedded image


wherein, Rcg is as previously defined.


In a further embodiment, group B is substituted by a substituent of Formula —NH2, —NH(RN1), —NH(RN2), wherein the —NH2, —NH(RN1), —NH(RN2) substituent is capped, by the replacement of a H atom, with a capping group selected from the group consisting of:




embedded image


In one embodiment, X1 is aspartyl or glutamyl and comprises a capping group. In one embodiment, X1 is aspartyl and comprises a capping group on the N-terminus. In one embodiment, X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is selected from the group consisting of:




embedded image


wherein, Rcg is as previously defined.


In a further embodiment, X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is selected from the group consisting of:




embedded image


In one embodiment, X1 is aspartyl and comprises a capping group on the N-terminus, wherein the capping group is selected from the group consisting of:




embedded image


wherein, Rcg is as previously defined.


In one embodiment, X1 is aspartyl and comprises a capping group on the N-terminus, wherein the capping group is selected from the group consisting of:




embedded image


Preferred capping groups are those selected from List 1:




embedded image


embedded image


In a further embodiment, group B is substituted by a substituent of Formula —NH2, —NH(RN1), —NH(RN2), wherein the —NH2, —NH(RN1), —NH(RN2) substituent is capped, by the replacement of a H atom, with a capping group selected from the group consisting of those selected from List 2:




embedded image


embedded image


In such an embodiment, X1 may be aspartyl or glutamyl, in particularly aspartyl, which comprises a capping group on the N-terminus, wherein the capping group is selected from the group consisting of List 2.


As described above, in some embodiments RN2 is —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids and comprises a capping group, wherein the capping group is selected from the group consisting of List 3:




embedded image


embedded image


In particularly Aa may be lysyl, with a capping group on an N as illustrated below (Formula M), in particular where the capping group is a capping group selected from List 3.




embedded image


Advantageously, the capping group on Aa does not have a detrimental effect on the activity of the compounds or modified peptides of the invention.


Where the compounds or modified peptides of the present invention include more than one capping group, all combinations of the capping groups described herein are envisaged. In particular, when group B has a capping group selected from List 2, and RN2 is as defined above in association with List 3, and has a capping group selected from List 3, all combinations of capping groups from List 2 and List 3 are envisaged.


Particular combinations of capping groups may increase the ability of the compounds and modified peptides of the invention to penetrate cells.


Exemplary combinations of capping groups are as follows;


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


and;


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


X1 is aspartyl or glutamyl and comprises a capping group on the N-terminus, wherein the capping group is:




embedded image


RN2 is, —(CH2)0-10-(Z7)0-1-Aa, wherein Z7 is C═O, and Aa is lysyl and comprises a capping group as illustrated in Formula M, wherein the capping group is;




embedded image


A capping group can be added to a compound according to any one of the above-described aspects of the invention. For example, the compound may be of Formula Id:





Capping group-X1-X2-X3-X4-X5-X6-X7   Formula Id


In one embodiment, the compound may be of Formula Ie





X1-X2-X3-X4-X5-X6-X7-Capping group   Formula Ie


In one embodiment, the compound may be of Formula If





Capping group-X1-X2-X3-X4-X5-X6-X7-Capping group   Formula If


In one embodiment, the compound may be a modified peptide of Formula Ig:





Capping group-X1-E/D/pS-G-X4-X5-E/D/pS-NHRN2   Formula Ig


In particular, the compound may be a modified peptide of Formula Iw:





Capping group-d-E-G-F(3F)—W-E-NHRN2


In particular, the compound may be a modified peptide of Formula Ix:





Capping group-d-E-G-F(3F)—W-E-NHRN2-Capping group


Cyclised Peptides


In one embodiment of the present invention, the compound may be a modified peptide, wherein said modified peptide may be cyclised.


Cyclisation of the modified peptide may require the addition of one or more additional residues to the peptide sequences described above. In particular, enough additional residues are required to enable a carboxy-terminal group at one end of the linear sequence to bind to the amino-terminal group at the other end of the sequence and form a cyclised peptide. 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 additional residues may be required for this purpose.


Chemical Groups


Halo


The term “halogen” (or “halo”) is used herein to refer to fluorine, chlorine, bromine and iodine. In one embodiment, “halogen” is fluorine. In another embodiment, “halogen” is chlorine.


Carbonyl and Carboxy


Structure C═O represents a carbonyl group, which is a carbon atom connected with a double bond to an oxygen atom, and tautomeric forms thereof. A carbonyl group may also be denoted as —C(O)—. Examples of moieties that contain a carbonyl include but are not limited to aldehydes —C(O)H, ketones —C(O)—(C1-C10 alkyl)-, carboxylic acids —CO2H, amides —C(O)NH2, —C(O)—NH(C1-C10 alkyl), —C(O)—N(C1-C10 alkyl)2, —NH—C(O)—(C1-C10 alkyl) and esters —C(O)—O(C1-C10 alkyl).


Amine, Amino Etc.


An amine group is denoted by —NH2, in which a nitrogen atom is covalently bonded to two hydrogen atoms. An alkylamino group is denoted by —NH(C1-C10 alkyl), in which a nitrogen atom is covalently bonded to one hydrogen atom and one (C1-C10 alkyl) group. A dialkylamino group is denoted by —N(C1-C10 alkyl)2, in which a nitrogen atom is bonded to at least two additional (C1-C10 alkyl) groups. Amines may be named in several ways. Typically, a compound is given the prefix “amino” or the suffix “amine”.


Alkyl, Cycloalkyl, Heterocyclyl, Alkenyl, Alkynyl


The term “alkyl” is used herein to refer to monovalent, divalent or trivalent straight or branched, saturated, acyclic hydrocarbyl groups. In one embodiment, alkyl is C1-C10 alkyl, in another embodiment C1-C6 alkyl, in another embodiment C1-C4 alkyl, such as methyl, ethyl, n-propyl, i-propyl, n-butyl or t-butyl groups.


The term “cycloalkyl” is used herein to refer to monovalent, divalent or trivalent saturated, cyclic hydrocarbyl groups. In one embodiment cycloalkyl is C3-10cycloalkyl, in another embodiment, C3-6cycloalkyl, such as cyclopentyl and cyclohexyl.


The term “heterocyclyl” is used herein to refer to monovalent, divalent or trivalent cycloalkyl groups in which up to three carbon atoms, in one embodiment up to two carbon atoms, in another embodiment one carbon atom, are each replaced independently by O, S(O)1-2 or N, provided at least one of the cycloalkyl carbon atoms remains.


Examples of heterocyclyl groups include oxiranyl, thiaranyl, aziridinyl, oxetanyl, thiatanyl, azetidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, 1,4-dioxanyl, 1,4-oxathianyl, morpholinyl, 1,4-dithianyl, piperazinyl, 1,4-azathianyl, oxepanyl, thiepanyl, azepanyl, 1,4-dioxepanyl, 1,4-oxathiepanyl, 1,4-oxaazepanyl, 1,4-dithiepanyl, 1,4-thieazepanyl and 1,4-diazepanyl. Other examples include cyclic imides, cyclic anhydrides and thiazolidindiones. The heterocyclyl group may be C-linked or N-linked, i.e. it may be linked to the remainder of the molecule through a carbon atom or through a nitrogen atom.


The term “alkenyl” is used herein to refer to monovalent, divalent or trivalent straight or branched, unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon double bond and, in one embodiment, no carbon-carbon triple bonds. In one embodiment, alkenyl is C2-C10 alkenyl, in another embodiment, C2-C6 alkenyl, in another embodiment C2-C4 alkenyl.


The term “alkynyl” is used herein to refer to monovalent or divalent unsaturated, acyclic hydrocarbyl groups having at least one carbon-carbon triple bond. In one embodiment alkynyl is C2-C10 alkynyl, in another embodiment, C2-C6 alkynyl, in another embodiment C2-C4 alkynyl.


Aryl


The term “aryl” is used herein to refer to monovalent, divalent or trivalent, aromatic, cyclic hydrocarbyl groups, such as phenyl or naphthyl (e.g. 1-naphthyl or 2-naphthyl). In general, the aryl group may be a monocyclic or polycyclic fused ring aromatic group. Preferred aryl groups are C6-C14aryl. Aryl groups include phenyl, biphenyl, naphthyl, indenyl, fluorenyl, anthracyl and phenanthryl.


Heteroaryl


The term “heteroaryl” is used herein to refer to monovalent, divalent or trivalent, heteroaromatic, cyclic hydrocarbyl groups additionally containing one or more heteroatoms independently selected from O, S, N and NRT, wherein RT is preferably H or C1-C10 alkyl. In general, the heteroaryl group may be a monocyclic or polycyclic fused ring heteroaromatic group. Examples of monocyclic heteroaromatic groups are pyridyl, thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazoyl, oxadiazolyl, thiadiazolyl and tetrazolyl. Examples of polycyclic heteroaromatic groups are indolyl, benzofuranyl, benzothienyl, quinolyl, isoquinolyl, indazolyl, indolinyl, isoindolyl, indolizinyl, benzimidazolyl, quinolinyl and isoquinolinyl. Further examples of heteroaromatic groups include:




embedded image


Isomeric Forms


Compounds of the invention may exist in one or more geometrical, optical, enantiomeric, diastereomeric and tautomeric forms, including but not limited to cis- and trans-forms, E- and Z-forms, R-, S- and meso-forms, keto-, and enol-forms. All such isomeric forms are included within the invention. The isomeric forms may be in isomerically pure or enriched form, as well as in mixtures of isomers (e.g. racemic or diastereomeric mixtures).


Exemplary Compounds


In one embodiment, the compounds of the invention comprise a sequence of amino acids according to the following Formula Ic:





X1-E/D/pS-G-X4-X5-E/D/pS-NHRN2   Formula Ic


In a further embodiment, a compound of the invention may be a modified peptide of Formula Ig:





Capping group-X1-E/D/pS-G-X4-X5-E/D/pS-NHRN2   Formula Ig


In one embodiment, the compound of the invention may be of Formula (IA)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


and


RA3, RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IAA)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3, RA4 and X1 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IAAA)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3, RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IAAAA)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3, RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IAAAAA)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3, RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IAAAAA), wherein each R4 is independently —CO2H, —CH2CO2H—OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3, RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IB)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IBB)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IBBB)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IBBBB)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IBBBBB)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of formula (IBBBBB), wherein each R4 is independently —CO2H, —CH2CO2H, —OP(O)(OH)2, triazole, tetrazole, sulfonamide or sulphate;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IC)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ICC)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ICCCC)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 selected from the group consiting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4. In one embodiment, the compound of the invention may be of Formula (ICCCC)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ICCCCC)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, NR1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ICCCCC) wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, NR1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2, and the caping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ID)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and X1 is as previously defined.


In one embodiment, the compound of the invention may be of Formula (IDD)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl; X1 is as previously defined and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IDDD)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl; X1 is as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group;


In one embodiment, the compound of the invention may be of Formula (IDDDD)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl; X1 is as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IDDDDD)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl; X1 is as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IDDDDD), wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl, X1 is as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3.


In one embodiment, the compound of the invention may be of Formula (IE)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and X1 is as previously defined.


In one embodiment, the compound of the invention may be of Formula (IEE)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; X1 is as previously defined and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IF)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and X1 is as previously defined.


In one embodiment, the compound of the invention may be of Formula (IFF)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; X1 is as previously defined and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IG)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined.


In one embodiment, the compound of the invention may be of Formula (IGG)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (IH)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and X1 is as previously defined.


In one embodiment, the compound of the invention may be of Formula (IHH)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; X1 is as previously defined and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (II)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (III)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJ)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;




embedded image


R3 is


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJJ)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJJJ)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJJJJ)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJJJJJ)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IJJJJJ), wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IK)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IKK)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IL)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (ILL)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IM)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IMM)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IMMM)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IMMMM)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl, RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IMMMMM)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IMMMMM), wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined, RN1 is as previously defined, Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IN)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 and X1 are as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (INN)




embedded image


wherein R4 is —CO2H, —CH2CO2H or —OP(O)(OH)2;




embedded image


R3 is


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 and X1 are as previously defined; and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IO)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 is as previously defined and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IOO)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 is as previously defined, RN1 and RN2 are as previously defined, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IOOO)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 is as previously defined, RN1 is as previously defined; Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IOOOO)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 is as previously defined, RN1 is as previously defined; Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IOOOO), wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2,


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; RA4 is as previously defined, RN1 is as previously defined; Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl, and CG is a capping group, wherein the capping group is selected from List 1. In one embodiment, the capping group on X1 is selected from List 2 and/or the capping group on Aa is selected from List 3. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IP)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is selected from the group consisting of —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 and C1-C10 alkyl; and RA4 is as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IQ)




embedded image


wherein each R4 is independently —CO2H, —CH2CO2H or —OP(O)(OH)2;


R3 is




embedded image


RA3 is —H, —F, —Cl, —Br, —I, —OH, —O(C1-C10 alkyl), —NO2 or C1-C10 alkyl; and RA4 is as previously defined. In one embodiment, R3 may be substituted at one or more positions with RA4.


In one embodiment, the compound of the invention may be of Formula (IS)




embedded image


wherein CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (ISS)




embedded image


wherein RN1 and RN2 are as previously defined, and CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (ISSS)




embedded image


wherein RN1 is as previously defined and CG is a capping group;


Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl.


In one embodiment, the compound of the invention may be of Formula (ISSSS)




embedded image


Wherein RN1 is as previously defined and CG is a capping group;


Aa is a chain of one or more non-naturally occurring amino acids, or a chain of one or more naturally occurring amino acids, or a chain of a mixture of one or more naturally occurring amino acids and one or more non-naturally occurring amino acids; wherein the one or more non-naturally occurring or naturally occurring amino acids are independently selected from the group consisting of L-amino acids, D-amino acids and aza-amino acids, in particular Aa is lysyl.


In one embodiment, the compound of the invention may be of Formula (ISSSSS)




embedded image


Wherein CG is a capping group.


In one embodiment, the compound of the invention may be of Formula (ISSSSS), wherein CG is a capping group selected from List 1. In one embodiment, the capping group on the “d” terminus is selected from List 2 and/or the capping group on the “K” terminus is selected from List 3.


In one embodiment, the compound of the invention may comprise or consist of a sequence selected from the group consisting of:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, any one or more of the residues included in the exemplary compounds described above may be an aza amino acid, wherein an “aza amino acid” is an L amino acid in which the α-carbon atom has been replaced by a nitrogen atom.


Prodrugs


In one embodiment, the compound may be formulated for administration to a patient as a prodrug. The term “prodrug” means a precursor of a designated compound that, following administration to a subject yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug, on being brought to physiological pH is converted to a compound of Formula Ia, Ib, Ic, Id, Ie, If or Ig). Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of prodrugs”, H. Bundgaard et al.


Prodrugs may be produced, for example, by derivatising free carboxylic acid groups of structures of Formula Ia, Ib, Ic, Id, Ie, If or Ig as amides or esters. In one embodiment, the prodrug is an alkyl, aryl, or heteroaryl ester of a compound of the invention. In another embodiment the prodrug may comprise or consist of a methyl, ethyl, propyl, butyl, pentyl, hexyl, or benzyl ester of a compound of the invention.


In a further embodiment, the prodrug may comprise a cycloalkyl ester, preferably a cyclopentyl ester of a compound of the present invention. In a further embodiment, the prodrug may comprise a —CO2CH2CH2(heterocyclyl) ester, wherein heterocyclyl is preferably morpholino, of a compound of the present invention. In a further embodiment, the prodrug may comprise a —C02(CH2CH2O)1-10—CH2CH3 (polyethylene glycol or PEG) ester of a compound of the present invention.


A prodrug may be a compound comprising an alcohol functionality, which when phosphorylated in vivo produces the active compound. For example, a compound comprising a serine residue may be a prodrug which, when subjected to physiological conditions is phosphorylated to form the corresponding phosphorylated serine residue, thereby producing the active compound.


Example of Prodrug



embedded image


Examples of PEG-Based Prodrugs



embedded image


embedded image


Methods of Manufacture


Compounds of the invention were synthesised by the coupling of smaller fragments/subunits, usually amino acids.


Amino acids that were commercially available were purchased and used directly (following any appropriate protecting group modification). Unnatural amino acids were synthesised starting from the appropriate amino acid precursor.


Carboxylic Acid Bioisostere Synthesis


Compounds of the present invention may comprise carboxylic acid bioisosteres. Such carboxylic acid bioisosteres may be synthesised by modification of the functionality of the side chain of an amino acid. Such functionality may be, for example, a carboxylic acid or amide. In this case, appropriate starting amino acids would include aspartic acid, glutamic acid, asparagine and glutamine. A representative scheme for the conversion of an amide functionality of the side chain of an amino acid into a tetrazole group is illustrated below (Tetrazole amino acids as competitive NMDA antagonists, Bioorganic & Medicinal Chemistry Letters, 1993):




embedded image


As will be appreciated by the skilled person, the scheme above is a representative procedure for the conversion of a natural amino acid into an unnatural amino acid.


Using standard synthetic procedures, the person skilled in the art would be able to synthesise other unnatural amino acids in an analogous manner to that shown above.


Once synthesised, the smaller fragments/subunits (usually amino acids —natural/unnatural) are coupled together to form compounds of the present invention.


The smaller fragments/subunits are coupled together using a solid-phase peptide synthesis. Reagents and conditions for this technique are illustrated in FIGS. 1A-1E.


Further experimental procedures are provided in the Examples section.


Pharmaceutical Compositions


The compound, modified peptide or prodrug of the invention may be formulated into a pharmaceutical composition. The invention therefore includes a pharmaceutical composition comprising one or more of the compounds, modified peptides or produgs of the invention.


In one embodiment the pharmaceutical composition may additionally comprise a pharmaceutically-acceptable carrier, excipient, diluent or buffer. Suitable pharmaceutically acceptable carriers, excipients, diluents or buffers may include liquids such as water, saline, glycerol, ethanol or auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like. Excipients may enable the pharmaceutical compositions to be formulated into tablets, pills, capsules, liquids, gels, or syrups to aid intake by the subject. A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences.


The pharmaceutical composition may include a therapeutically effective amount of one or more of the compounds, modified peptides or produgs of the invention. A pharmaceutically effective amount is an amount able to treat the disease for which the composition is intended. The actual amount will depend on a number of factors including the size, weight, age, gender, and health of an individual, and the rate of blood clearance, and will be decided by a clinical practitioner. Generally a pharmaceutically effective amount will be between 1 g/kg body weight and 1 mg/kg body weight or less.


In one embodiment the pharmaceutical composition may include an additional pharmaceutically active agent such as a therapeutic component, in particular, a component useful for the treatment of hyperproliferative disorders such as cancer, inflammatory disorders involving the NFkB signalling pathway such as arthritis, osteoarthritis, rheumatoid arthritis, Crohn's Disease and Irritable Bowel Syndrome (IBS), infectious disorders or neurodegenerative disorders and may include chemotherapeutics, ERMs, SERMs, other E1, E3, E3 and deubiquitinating enzyme inhibitors, proteasome inhibitors, kinase inhibitors, HDAC inhibitors, PPAR inhibitors or specific biological targeted therapies e.g. Herceptin.


The invention also includes any medical device which may have the pharmaceutical composition of the invention inserted into it or coated onto it. Such devices include but are not limited to stents, pins, rods, meshes, beads, syringes, plasters, microchips, micro fluidic devices, and stitches.


Methods of Treatment


In another aspect, the invention includes a compound, modified peptide, prodrug or pharmaceutical composition of the invention for use in medicine.


In one embodiment the invention provides a compound, modified peptide, prodrug or pharmaceutical composition of the invention for use in the treatment of a disease associated with aberrant protein degradation.


In one embodiment the invention provides a compound, modified peptide, prodrug or pharmaceutical composition of the invention for use in the treatment of a hyperproliferative disorder such as cancer, inflammatory disorders involving the NFkB signalling pathway such as arthritis, osteoarthritis, rheumatoid arthritis, Crohn's Disease and Irritable Bowel Syndrome (IBS), infectious disorders or neurodegenerative disorders.


In another embodiment the invention includes a method of treating a hyperproliferative disorder such as cancer, inflammatory disorders involving the NFkB signalling pathway such as arthritis, osteoarthritis, rheumatoid arthritis, Crohn's Disease and Irritable Bowel Syndrome (IBS), infectious disorders or neurodegenerative disorders comprising administering a pharmaceutically effective amount of a compound, modified peptide, prodrug or pharmaceutical composition of the invention to a patient in need of treatment.


In a particular embodiment, the invention includes a method of treating breast cancer or prostate cancer comprising administering a pharmaceutically effective amount of a compound, modified peptide, prodrug or pharmaceutical composition of the invention to a patient in need of treatment.


As used herein, the term “treatment” encompasses therapy, and can be prophylactic or therapeutic.


A pharmaceutically effective amount is an amount able to treat the disease for which the compound, modified peptide, prodrug or pharmaceutical composition has been administered. The actual amount will depend on a number of factors including the size, weight, age, gender, health of an individual, and the rate of blood clearance, and will be decided by a clinical practitioner. Generally a pharmaceutically effective amount will be between 1 g/kg body weight and 1 mg/kg body weight or less.


In another embodiment the invention includes the use of a compound, modified peptide, prodrug or pharmaceutical composition of the invention in the manufacture of a medicament for the treatment of a hyperproliferative disorder such as cancer, inflammatory disorders involving the NFkB signalling pathway such as arthritis, osteoarthritis, rheumatoid arthritis, Crohn's Disease and Irritable Bowel Syndrome (IBS), infectious disorders or neurodegenerative disorders.


In a particular embodiment, the invention includes the use of a compound, modified peptide, prodrug or pharmaceutical composition of the invention in the manufacture of a medicament for the treatment of breast cancer or prostate cancer.


The compound, modified peptide, prodrug or pharmaceutical composition of the invention may be used for the treatment of disease in any animal. The animal may be a mammal such as a camel, dog, cat, horse, cow, pig, sheep, camelid, mouse, rat, rabbit, hamster, guinea pig, pig, or sheep. In one embodiment, the mammal may be a human.


The compound, modified peptide, prodrug or pharmaceutical composition of the invention may be administered to a patient using any one or more of a number of modes of administration which will be known to a person skilled in the art. Such modes of administration may include parenteral injection (e.g. intravenously, subcutaneously, intraperitoneally, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral, vaginal, topical, transdermal, intradermal, intrathecal, intranasal, ocular, aural, pulmonary or other mucosal administration. The precise mode of administration will depend on the disease or condition to be treated.


Diagnostic Kits


In another aspect, the invention includes a diagnostic kit comprising a compound, modified peptide or prodrug of the invention. Within this aspect, the compound, modified peptide or prodrug may be labelled to allow its identification. Suitable labels may include, coloured labels, fluorescent labels, and radioactive labels. Detection may be performed by FACS, Western blot, immunoblot or any other technique known to be useful for the identification of labelled molecules.


Diagnostics kits may be used to identify patients having increased βTrCP expression.


As discussed above, increased βTrCP expression can be associated with aberrant protein degradation mechanisms, which can lead to hyperproliferative disorders such as cancer through the increased degradation of pro-apoptotic factors. Increased βTrCP expression can also lead to inflammatory disorders involving the NFkB signalling pathway such as arthritis, osteoarthritis, rheumatoid arthritis, Crohn's Disease and Irritable Bowel Syndrome (IBS), infectious disorders and neurodegenerative disorders.


Diagnostics kits may also comprise instructions.


Various aspects and embodiments of the present invention will now be described in more detail by way of example. It will be appreciated that modification of detail may be made without departing from the scope of the invention.





BRIEF DESCRIPTION OF FIGURES


FIGS. 1A, 1B, and 1C show solid supported peptide synthesis.


Reagents and Conditions: a) Rink amide linker (3 equiv), oxyma (3 equiv), DIC (3 equiv), 0.1 M in DMF, 30 min; b) 20% piperidine in DMF (2×5 min); c) Amino acid (3 equiv), HBTU (3 equiv), DIPEA (6 equiv) 0.1 M in DMF, 40 min; d) TsCl (5 equiv), DMAP (0.1 equiv), DIPEA (10 equiv), 0.1 M in DMF, 40 min; e) TFA, 5% TIS, 5% DCM, 3h.



FIGS. 1D and 1E show solid supported peptide synthesis for C-terminal modified peptides.


Reagents and Conditions: a) Rink amide linker (3 equiv), oxyma (3 equiv), DIC (3 equiv), 0.1 M in DMF, 30 min; b) 20% piperidine in DMF (2×5 min); c) Amino acid (3 equiv), HBTU (3 equiv), DIPEA (6 equiv) 0.1 M in DMF, 40 min; d) TsCl (5 equiv), DMAP (0.1 equiv), DIPEA (10 equiv), 0.1 M in DMF, 40 min; e) 2% Hydrazine in DMF (6×15 mins); f) BzCl (5 equiv), DMAP (0.1 equiv), DIPEA (10 equiv), 0.1 M in DMF, 40 min; g) TFA, 5% TIS, 5% DCM, 3h.



FIG. 2 shows the abbreviations used to represent capping groups used in synthesis.



FIG. 3 shows the abbreviations used to represent acidic capping groups.



FIGS. 4A and 4B show the abbreviations used to represent non-natural amino acids.



FIGS. 5A, 5B, and 5C show FP assay dose response curves. Peptides are numbered according to Table 11.



FIG. 6 shows Biotin pulldown assay results. Peptides are numbered according to Table 11.



FIG. 7 shows SPR assay results. FIG. 7A) Ts-DEGF(3C1)W-(Me)E-NH2 (SEQ ID NO: 118); FIG. 7B) TsdEGF(3C1)WE-NH2; FIG. 7C) 4-(MeO)-PhS02-dEGF(3F)WE-NH2; FIG. 7D) Ts-DEGF(3F)WENH2 (SEQ ID NO: 3); and FIG. 7E) Ts-dEGF(3F)W-(Me)E-NHz. Peptides are numbered according to Table 11.



FIG. 8 shows ubiquitination assay results. Peptides are numbered according to Table 11.



FIGS. 9A and 9B show peptidomimetic selectivity vs other E3s. Peptides are numbered according to Table 11.



FIG. 10 shows blots of immunoprecipitated proteins from HeLa cells transfected with βTrCP and substrates and treated with cell-permeable βTrCP disruptor peptides.


Peptides are numbered according to Table 11: DSGIFE (SEQ ID NO: 119); DpSGIFE (SEQ ID NO: 12); and succ-EGFFE (SEQ ID NO: 57).



FIGS. 11A and 11B show the ELSDs, which shows the mass of the desired peptide. Peptides are numbered according to Table 11.



FIG. 12 shows the accumulation of PDCD4 following nucleofection with the peptide 4-(MeO)-PhSO2-dEGF(3F)WE-NH2 observed using an in cell Western assay, expressed as % activity.



FIG. 13 shows the accumulation of GFP-PDCD4 following nucleofection with the peptide 4-(MeO)-PhSO2-dEGF(3F)WE-NH2 observed using a fluorescent reporter assay, expressed as % activity.



FIGS. 14A and 14B show the collation of the assay results for the cell permeable compounds.



FIG. 15 shows the accumulation of PDCD4 in MCF7 cells as measured by in cell western assay following treatment with UBP036.



FIG. 16 shows the activity of round II compounds in relation to UBP036.



FIGS. 17a and 17b show accumulation of β-catenin in MCF7 cells following treatment with UBP036 measured by in cell western assay.



FIGS. 18a and 18b show GFP-PDCD4 accumulation in MCF7 cells following treatment with UBP036.



FIG. 19 shows PDCD4 accumulation in MCF7 cells following treatment with UBP036, UBP037 and UBP038 measured by traditional western blot.



FIGS. 20a and 20b show PDCD4 accumulation in LNCaP cells following treatment with UBP036, UBP037 and UBP038.



FIG. 21 shows cell viability of MCF7 cells following treatment with UBP036 measured on the xCELLigence platform.



FIGS. 22A, 22B, 22C, 22D, and 22E show cell viability following compound treatment as measured by the xCELLigence platform (A) cell proliferation of UBP036, UBP037 and UBP038 at 20 uM; (B, D, E) dose response curve for UBP036, UBP037 and UBP038; (C) cell proliferation of UBP036 compared to the control compound.



FIG. 23 shows the inhibition of cancer cell growth compared to non-cancer cell growth following treatment with UBP036, UBP037 and UBP038.



FIG. 24 shows PDCD4 accumulation following nucleofection of UBP022 into MCF7 cells.





EXAMPLES
Experimental Procedures

The following experimental conditions were used throughout the examples unless other details are provided.


General Conditions for the Solid-Phase Synthesis


All the coupling reactions were carried out at room temperature if no specifications are given. Solid-phase synthesis was performed manually using Isolute filtration reservoirs as the reaction vessel, fitted with polyethylene frits (Argonaut Technologies Inc). Amino acids are Fmoc protected at the N terminus, with suitable acid labile protecting groups on the side chains. For C-terminal modified peptides Fmoc-Lys(Dde)-OH was used to allow selective modification of the Lys side chain. Each coupling step of the synthesis was assessed for completion using either the Kaiser test for primary amines, or the chloranil test for secondary amines.


Coupling the Linker to the Resin


Aminomethyl PS resin (loading 1.23 mmol/g, 0.30 g, 0.369 mmol) in a 6 mL reaction vessel was swollen for 5 minutes in DCM (3 mL), then washed with DCM (3×3 mL). To a solution of Rink amide linker (598 mg, 1.11 mmol) in DMF (3.69 mL) was added oxyma (157 mg, 1.11 mmol) and the solution shaken for 10 minutes. DIC (173 uL, 1.11 mmol) was added and the solution shaken for 2 minutes. The mixture was added to the resin and shaken for 30 minutes. The resin was filtered and washed with DMF (3×4 mL), DCM (3×4 mL) and MeOH (3×4 mL). Kaiser test negative. The resin was washed with Et2O (3×4 mL) and dried under vacuum for storage.


Coupling of Amino Acids/Spacer


Resin (˜0.049 mmol) in a 3 mL reaction vessel was swollen for 5 minutes in DCM (1.5 mL) and filtered. A solution of 20% piperidine in DMF (1.5 mL) was added, the vessel was shaken for 5 mins and the resin was filtered and washed with DMF (3×1.5 mL) and DCM (3×1.5 mL). Piperidine deprotection and washing cycle was repeated and the resin was dried under vacuum, Kaiser test positive. To a solution of the appropriate amino acid/spacer (0.15 mmol, 3 equiv) in DMF (0.49 mL) was added HBTU (0.15 mmol, 3 equiv) and the solution shaken for 2 minutes. DIPEA (0.30 mmol, 6 equiv) was added and the solution shaken for 1 minute. The mixture was added to the resin and shaken for 40 minutes. The resin was filtered and washed with DMF (3×1.5 mL), DCM (3×1.5 mL) and MeOH (3×1.5 mL). Kaiser test negative, otherwise treatment of activated amino acid repeated.


Coupling to N-Alkylated Amino Acids


Resin (˜0.049 mmol) in a 3 mL reaction vessel was swollen for 5 minutes in DCM (1.5 mL) and filtered. A solution of 20% piperidine in DMF (1.5 mL) was added, the vessel was shaken for 5 mins and the resin was filtered and washed with DMF (3×1.5 mL) and DCM (3×1.5 mL). Piperidine deprotection and washing cycle was repeated and the resin was dried under vacuum, Choranil test positive. To a solution of the appropriate amino acid (0.15 mmol, 3 equiv) in DMF (0.49 mL) was added oxyma (0.15 mmol, 3 equiv) and the solution shaken for 10 minutes. DIC (0.15 mmol, 3 equiv) was added and the solution shaken for 2 minutes. The mixture was added to the resin and heated in a microwave at 60° C. for 20 minutes. The mixture was then shaken for an additional 20 minutes. The resin was filtered and washed with DMF (3×4 mL), DCM (3×4 mL) and MeOH (3×4 mL). Chloranil test negative, otherwise treatment of activated amino acid repeated.


Example of the N-Terminus Capping


Resin (˜0.049 mmol) in a 3 mL reaction vessel was swollen for 5 minutes in DCM (1.5 mL) and filtered. A solution of 20% piperidine in DMF (1.5 mL) was added, the vessel was shaken for 5 mins and the resin was filtered and washed with DMF (3×1.5 mL) and DCM (3×1.5 mL). Piperidine addition and washing cycle was repeated and the resin was dried under vacuum, Kaiser test positive. To a solution of 4-toluenesulfonyl chloride (0.25 mmol, 5 equiv) in DCM:DMF (1:1, 0.49 mL) was added DMAP (0.005 mmol, 0.1 equiv) and the solution shaken for 2 minutes. DIPEA (0.50 mmol, 10 equiv) was added and the solution shaken for 1 minute. The mixture was added to the resin and shaken for 40 minutes. The resin was filtered and washed with DMF (3×1.5 mL), DCM (3×1.5 mL) and MeOH (3×1.5 mL). Kaiser test negative, otherwise treatment of with the capping group was repeated.


Example of the C-Terminus Capping


After N-terminus capping, resin (˜0.049 mmol) in a 3 mL reaction vessel was swollen for 5 minutes in DCM (1.5 mL) and filtered. A solution of 2% hydrazine monohydrate in DMF (1.5 mL) was added, the vessel was shaken for 15 mins and the resin was filtered and washed with DMF (3×1.5 mL) and DCM (3×1.5 mL). Hydrazine addition and washing cycle was repeated (×5) and the resin was dried under vacuum, Kaiser test positive. To a solution of benzoyl chloride (0.25 mmol, 5 equiv) in DCM:DMF (1:1, 0.49 mL) was added DMAP (0.005 mmol, 0.1 equiv) and the solution shaken for 2 minutes. DIPEA (0.50 mmol, 10 equiv) was added and the solution shaken for 1 minute. The mixture was added to the resin and shaken for 40 minutes. The resin was filtered and washed with DMF (3×1.5 mL), DCM (3×1.5 mL) and MeOH (3×1.5 mL). Kaiser test negative, otherwise treatment of with the capping group was repeated.


Characterization of selected examples:















ID





(SEQ ID


RT


NO:)
Structure
MS
(mins)


















UBP001
DpSGIFE-NH2
744.1a
4.272e


(12)


UBP002
Suc-EGFFE-NH2
725.2a
5.107e


(57)


UBP003
Suc-EGF(2F)F(4NO2)E-NH2
788.2a
4.989e


(58)


UBP004
Suc-EGF(3F)F(4NO2)E-NH2
788.2a
5.073e


(58)


UBP005
Suc-EGF(4F)F(4NO2)E-NH2
788.2a
5.088e


(58)


UBP006
Suc-EGF(2F)Y(Me)E-NH2
773.2a
5.023e


(59)


UBP007
Suc-EGYFE-NH2
741.1a
4.500e


(60)


UBP008
Mal-EGF(3F)F(4NO2)E-NH2
786.1a
3.739e


(61)


UBP009
Suc-EGY1NalE-NH2
791.2a
5.222e


(62)


UBP010
Suc-EGF(3F)1NalE-NH2
793.0a
5.783e


(62)


UBP011
Suc-EGF(4NO2)1NalE-NH2
820.0a
5.775e


(62)


UBP012
Suc-QGF(3F)F(4NO2)E-NH2
787.0a
4.924e


(63)


UBP013
Suc-EGYF(4NO2)E-NH2
786.1a
4.422e


(64)


UBP014
Suc-EGF(3F)WE-NH2
781.9a
4.597e


(65)


UBP015
Suc-EGF(3F)HE-NH2
733.0a
3.257e


(65)


UBP016
Ac-dEGF(3F)1NalE-NH2
850.0a
5.705e


UBP017
Ac-dEGF(3F)WE-NH2
838.9a
4.976e


UBP018
Bz-dEGF(3F)WE-NH2
901.0a
5.514e


UBP019
Et(CO)-dEGF(3F)WE-NH2
853.0a
5.167e


UBP020
MeO(CO)-dEGF(3F)WE-NH2
855.1a
5.138e


UBP021
Ts-dEGF(3F)WE-NH2
951.2a
5.800e


UBP022
4-(MeO)-PhSO2-dEGF(3F)WE-NH2
967.2a
5.690e


UBP023
EtO(CO)-dEGF(3F)WE-NH2
896.2a
5.362e


UBP024
Ts-DEGF(3F)WE-NH2
951.2a
5.673e


(66)


UBP025
Ts-dDGF(3F)WE-NH2
937.2a
5.745e


UBP026
4-(MeO)-PhSO2-DEGF(3F)WE-NH2
967.2a
5.534e


(67)


UBP027
Ts-dEGF(3F)WD-NH2
937.2a
5.770e


UBP028
Ts-dDGF(3F)WD-NH2
923.2a
5.729e


UBP029
3,4-(MeO)2-PhSO2-dEGF(3F)WE-NH2
997.2a
5.495e


UBP030
4-(BuO)-PhSO2-dEGF(3F)WE-NH2
1009.4a
6.404e


UBP031
2-NaphthylSO2-dEGF(3F)WE-NH2
987.2a
6.010e


UBP032
Ts-dEGF(3F)WE(Me)-NH2
956.2a
5.891e


UBP033
Ts-dEGF(3Cl)WE(Me)-NH2
981.2a
5.992e


UBP034
Ts-dEGF(3Cl)WE-NH2
967.2a
6.086e


UBP035
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-kkkkkkkkk-NH2
2236.2b
3.547e


UBP036
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(Ahx-Chol)-NH2
1773.4d
7.970f


UBP037
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCOC17H35)-NH2
1499.0c
10.398f


UBP038
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCOC19H39)-NH2
1527.2c
11.247f


UBP039
4-(t-Bu)-PhSO2-dEGF(3F)WE-NH2
1017.0c
9.502g


UBP040
4-(i-Pr)-PhSO2-dEGF(3F)WE-NH2
1003.1a
9.688g


UBP041
4-(Pr)-PhSO2-dEGF(3F)WE-NH2
1003.1c
9.471g


UBP042
4-(Br)-PhSO2-dEGF(3F)WE-NH2
1039.2c
8.589g


UBP043
4-(Br)-2-(CH3)-PhSO2-dEGF(3F)WE-NH2
1053.0c
8.932g


UBP044
2-Naph-SO2-dEGF(3F)WE-NH2
1011.2c
8.924g


UBP045
4-(OCF3)-PhSO2-dEGF(3F)WE-NH2
1045.0c
9.289g


UBP046
4-(Br)-3-(CF3)-PhSO2-dEGF(3F)WE-NH2
1107.0c
9.866g


UBP047
4-(CF3)-PhSO2-dEGF(3F)WE-NH2
1029.2c
9.319g


UBP048
2,4-(Cl)2-PhSO2-dEGF(3F)WE-NH2
1029.0c
9.437g


UBP049
2,4-(Br)2-PhSO2-dEGF(3F)WE-NH2
1117.0c
9.042g


UBP050
3,5-(CH3)2-PhSO2-dEGF(3F)WE-NH2
989.0c
8.959g


UBP051
4-(Br)-2-(OCF3)-PhSO2-dEGF(3F)WE-NH2
1123.0c
9.499g


UBP052
4-(I)-PhSO2-dEGF(3F)WE-NH2
1096.5c
11.312g


UBP053
4-(Cl)-PhSO2-dEGF(3F)WE-NH2
995.2c
10.944g


UBP054
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(t-Bu)-Ph))-NH2
1392.5c
10.163g


UBP055
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-2-Naph)-NH2
1386.3c
9.834g


UBP056
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(2,4,6-(Me)3-Ph))-NH2
1378.5c
9.313g


UBP057
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Me)-Ph))-NH2
1350.2c
9.462g


UBP058
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Br)-Ph))-NH2
1414.3c
9.497g


UBP059
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-(Br)-Ph))-NH2
1450.2c
9.903g


UBP060
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Cl)-Ph))-NH2
1370.3c
9.289g


UBP061
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCOPh)-NH2
1335.6c
10.507g


UBP062
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(3,5-(Cl)2-Ph))-NH2
1380.0a
10.035g


UBP063
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(CH2)4CH3)-NH2
1330.6c
9.493g


UBP064
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(CF3)-Ph))-NH2
1380.3a
9.874g


UBP065
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCOO-Ph)-NH2
1352.3c
9.575g


UBP066
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(OMe)-Ph))-NH2
1366.2c
8.875g


UBP067
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCONH-Ph)-NH2
1327.4a
9.389g


UBP068
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-CH2CH(CH3)2)-NH2
1316.0c
8.316g


UBP069
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHOCO-1-Naph)-NH2
1378.2a
10.118g


UBP070
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Cl)-2,6(F)2-Ph))-NH2
1406.5c
10.815g


UBP071
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Me2N)-Ph))-NH2
1379.3c
8.889g


UBP072
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-(i-Pr)-Ph))-NH2
1414.0c
10.271g


UBP073
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2Ph)-NH2
1348.2a
8.877g


UBP074
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-(n-Pr)-Ph)-NH2
1414.2c
10.269g


UBP075
4-(t-Bu)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(t-Bu)Ph))-NH2
1394.3a
7.138e


UBP076
4-(t-Bu)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2-Naphth))-NH2
1388.2a
6.922e


UBP077
4-(t-Bu)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2,4,6-(Me)3-Ph))-NH2
1380.3a
6.911e


UBP078
4-(t-Bu)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Me)Ph))-NH2
1352.3a
6.802e


UBP079
4-(t-Bu)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Br)Ph))-NH2
1418.2a
6.917e


UBP080
4-(i-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(t-Bu)Ph))-NH2
1380.4a
7.037e


UBP081
4-(i-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2-Naphth))-NH2
1374.2a
6.813e


UBP082
4-(i-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2,4,6-(Me)3Ph))-NH2
1366.3a
6.786e


UBP083
4-(i-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO((4-(Br)Ph))-NH2
1402.2a
6.815e


UBP084
4-(n-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(tBu)Ph))-NH2
1380.3a
7.175e


UBP085
4-(n-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2-Naphth))-NH2
1374.3a
6.853e


UBP086
4-(n-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO((2,4,6-(Me)3Ph))-NH2
1366.3a
6.826e


UBP087
4-(n-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Me)Ph))-NH2
1338.3a
6.735e


UBP088
4-(n-Pr)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Br)Ph))-NH2
1404.2a
6.858e


UBP089
4-(Br)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(tBu)Ph))-NH2
1418.2a
7.047e


UBP090
4-(Br)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2-Naphth))-NH2
1412.2a
6.848e


UBP091
4-(Br)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2,4,6-(Me)3Ph))-NH2
1404.2a
6.814e


UBP092
4-(Br)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Me)Ph))-NH2
1376.1a
6.732e


UBP093
4-(Br)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Br)Ph))-NH2
1442.1a
6.866e


UBP094
4-(Br)-2-(Me)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(tBu)Ph))-NH2
1432.2a
7.036e


UBP095
4-(Br)-2-(Me)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2-Naphth))-NH2
1426.3a
6.831e


UBP096
4-(Br)-2-(Me)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(2,4,6-(Me)3Ph))-NH2
1418.2a
6.805e


UBP097
4-(Br)-2-(Me)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Me)Ph))-NH2
1390.2a
6.696e


UBP098
4-(Br)-2-(Me)PhSO2-dEGF(3F)WE-Ahx-K(NHCO(4-(Br)Ph))-NH2
1452.0a
6.824e






aMass identified as [M − H];




bMass identified as [M + H]+;




cMass identified as [M + Na]+;




dMass identified as [M + K]+;



HPLC analysis preformed using a Supleco Discovery C18 5 cm × 4.6 mm, 5 μm column, samples analysed by ELSD, 220 nM and 254 nM, conditions used where e5% to 95% MeOH (+0.1% formic acid) in H2O (+0.1% formic acid) over 6 minutes, 3 minute hold, then 1 minute at 5% MeOH (+0.1% formic acid);



f5% to 95% MeCN (+0.1% formic acid) in H2O (+0.1% formic acid) over 10 minutes, 4 minute hold, then 1 minute at 5% MeCN (+0.1% formic acid);




g5% to 95% MeOH (+0.1% formic acid) in H2O (+0.1% formic acid) over 10 minutes, 4 minute hold, then 1 minute at 5% MeOH (+0.1% formic acid);







Cleavage from Resin


Resin (˜0.0.49 mmol) in a 3 mL reaction vessel was swollen for 5 minutes in DCM (2 mL) and filtered. A solution of TFA:TIS:DCM (90:5:5 0.49 mL) was added, and the vessel was shaken for 3 h. The resin was removed by filtration, and ice-cold Et2O (10 mL) was added to the filtrate. The resultant solid was pelleted by centrifuge, and the solvent removed by decantation. Solid was dried under vacuum.


The experimental scheme for solid phase synthesis is shown in FIGS. 1A-1E.


Fluorescence Polarization Screening of βTrCP


Assay Components


0.035 μM βTrCP (tag cleaved and complexed with Skp1)


10 nM fluorescein-RHDpSGLDpSMKD (SEQ ID NO:68)


50 mM Hepes pH 7.5


50 mM NaCl


1 mM DTT


0.1 mg/ml BSA (Bovine Serum Albumin)


50 μM compound (in DMSO)


Assay Protocol


Assay components (without compound) were premixed in a microcentrifuge tube and incubated for 1 hour to ensure equilibrium was achieved. Each compound was then added to one tube, mixed by vortexing, and then dispensed into 3 wells of a black 384-well plate and incubated for 30 minutes. Fluorescence polarization was then read (excitation 485 nM, emission 530 nM) using an Analyst-AD from Molecular Devices.


For dose-response curves to determine Ki, 10 different concentrations of compound were tested at equally spaced intervals. DMSO was added such that final concentration was 2%. Conditions are very tolerant to DMSO. Up to 10% DMSO has been tested previously, with no significant change to Kd values.


Surface Plasmon Resonance (SPR)


Experiments were carried out using the Biacore T200 SPR detection system. This system exploits the phenomenon of surface plasmon resonance (SPR) to monitor interactions between molecules. The system involves the attachment of one interacting partner to a surface (an appropriate sensor chip) while the other interacting partner is passed over it in solution. The binding of molecules to the surface generates an SPR response (measured in response units (RU)) that is proportional to the mass and amount of the biomolecule (in this case βTrCP/Skp1) bound to the chip. The relative responses obtained are dependent on the concentration of the molecule binding. At RUmaximum, the attached protein's binding sites are saturated. Binding events can be followed in real time and a range of interaction characteristics can be determined including kinetics, specificity of interactions and the concentration of specific molecules in a sample.


As βTrCP was His tagged, an NTA sensor chip was used. This sensor chip has a dextran surface matrix with immobilized nitrilotriacetic acid (NTA) which provides a means of capturing polyHis-tagged proteins through Nickel chelation. It was hoped that addition of Ni+ would orient the protein in a specific (and hopefully active) manner as it is covalently immobilised via amine coupling. Addition of EDC:NHS (N-ethyl-N′-(3-diethylaminopropyl)-carbodiimide:N-hydroxysuccinimide) converts carboxyl groups on the dextran sensor chip surface to succinamide esters which readily form covalent bonds with primary amines. Each chip contains four flow cells (each a separate surface) which means that compounds/peptides can be passed over different forms of βTrCP and a reference surface simultaneously. If binding to βTrCP is occurring, responses should be the same (accounting for differences in density of surface etc) on each surface.


Two different βTrCP protein complexes were immobilised on to an NTA biacore sensor chip. One included the GSTSkp1 fusion (HisβTrCP/GSTSkp1) while the other was immobilised after GST Removal by Thrombin (HisβTrCP/Skp1). To ensure that the GST moiety is completely removed from Skp1, βTrCP/GSTSkp1 was incubated with thrombin ((10units/mg protein) for at least 16 hours at room temperature in 10mMHEPES 150mMNaCl pH7.4+2 mM CaCl2. Thrombin and GST were removed from βTrCP/Skp1 by buffer exchange through a 50 KDa MWCO vivaspin concentrator.


Protein Immobilisation Procedure for NTA Chip


Ni+ (500 μM NiCl) loaded on to surface at a flow rate of 5 μl/min for 60s. EDC/NHS (activates dextran carboxylates) loaded at a flow rate of 5 μl/min for 240s. Protein ([protein]=100 nM to 1 μM) loaded at a flow rate of 10 μl/min for 180s. Strip solution (350 μM EDTA/1M NaCl) added at flow rate of 10 μl/min for 30s (to chelate excess Ni+ and remove non-covalently bound protein e.g. protein oligomers). Quench solution (ethanolamine) at flow rate of 5 μl/min for 240s (to deactivate surface molecules on the chip that have not crosslinked protein). The immobilisation buffer used was 10 mM HEPES pH 7.4, 150 mM NaCl,


GST Capture Procedure (for Immobilisation Via GSTSkp1)


EDC:NHS was injected at 5 μl/min for 4 min to activate surface for amine coupling (this converts carboxyl groups on the surface of the chip to succinamide esters that react with primary amines)


Anti-GST (60 μg/ml). was loaded at 10 μl/min for 4 min resulting in an increase in response units of 6730 (Biacore manual states it should result in ˜7000)


Ethanolamine was injected at 5 μl/min for 5 min to deactivate remaining unreacted esters at surface (quenching).


Injection of a low concentration of purified GST (from kit) was injected for 3 mins at 5 μl/min before running a regeneration cycle with glycine pH2.0 that disrupts the antibody-GST interaction. This step is recommended in the Biacore manual in order to “block” a minority of high affinity GST binding sites that may prevent regeneration and therefore reloading of fresh GST-protein of interest.


GSTSkp1/βTrCP (0.16 mg/ml) was then loaded at 10 μl/min for 4 min resulting in an increase of 1550 RU (2000 RU is about the maximum to expect according to Biacore manual).


SPR Assay Conditions


Small molecule/peptide samples to be assayed for binding to βTrCP surfaces, were provided as 10 mM stocks in 100% DMSO. All samples were tested in running buffer composed of 10 mM HEPES pH 7.4, 150 mM NaCl, 50 μM EDTA, 0.005% p20, and 1% DMSO. Serial dilutions were made using running buffer. Samples were tested over varying concentrations up to a maximum of 100 μM. Two methods of measuring the SPR response were employed: single cycle kinetics which measures the response across different concentrations of sample within a single cycle (no regeneration of surface) and a method that measures the response at a given concentration in each cycle and includes a regeneration wash after each sample injection. The regeneration solution used was the same as running buffer, but included 500 mM NaCl. Data was fitted using Biacore T200 evaluation software. KD values calculated from binding curves from both surfaces (with or without the GST moiety) were averaged to produce apparent KD's for each sample tested.


Biotin Pull Down Assay


Assay components


150 mM NaCl


10.01% NP40


1 mM DTT


0.3 μM βTrCP (1 μg)


0.3 μM biotinylated IκB peptide (KKERLLDDRHDpSGLDpSMKDEE)


100 μM compound (mabridge library: 50 μM)


Protocol


βTrCP1 and biotinylated peptide were incubated in a volume of 25 μl at a final concentration of DMSO of 1% for 30 minutes to achieve equilibrium. Compounds were then added to a final concentration of 100 μM and allowed to incubate for an additional 30 minutes. 7.5 μl of streptavidin-agarose beads were then added to the reaction mix and allowed to incubate at room temperature for 30 minutes with gentle rocking. Beads were spun down and washed in buffer 3 times and then loaded onto a 10% SDS PAGE gel and visualized by GelCode blue staining.


βTrCP Ubiquitination Assay and Selectivity Assays


Assay Components


0.2 μM E1 (Ube1)


2 μM E2 (UbcH5C)


0.25 μM E3 (Cul1/Rbx1)


0.25 μM E3 (β-TrCP1/Skp1)


12 μM Ubiquitin


0.5 μM Peptide substrate (biotin)


10 mM MgCl2


2 mM ATP


Protocol


Master mixes were prepared in a 50 mM Herpes buffer at pH 7.5, in 75 mM NaCl and 1 mM DTT without Mg or ATP and peptides were added to a final concentration of 100 μM. Reactions were incubated at room temperature for 30 minutes and then Mg/ATP was added to the mix. Reactions were further incubated for an additional 60 minutes and then stopped by adding SDS gel loading buffer and boiled for 5 minutes. Reactions were run on SDS-PAGE (10%) and transferred to nitrocellulose membranes and probed with HRP-Streptavidin.


FBW7 selectivity assays were performed in the same manner except using FBW7/Skp1 as E3 component and cyclin E as the substrate. Blots were probed using anti-cyclin E antibody.


EXAMPLES

The following Examples illustrate the experiments performed by the inventors to arrive at the preset invention. It will be appreciated that modification of detail may be made without departing from the scope of the invention.


Example 1 Construction of Binding Peptides and Analysis Using Fluorescence Polarisation (FP) Assay

A consensus binding motif is known to be present in IkBa, Vpu and β-catenin, all of which bind βTrCP (J. Pons et al., Biochemistry, 2008, 47 (1), 14-29). The consensus motif has the sequence DpSGXXpS (SEQ ID NO:69), wherein the two serine residues are phosphorylated.


A selection of compounds were prepared using the synthesis procedure described above, and shown in FIGS. 1A-1E. Here, each residue of the consensus binding motif was replaced in turn as shown in Table 1.


Binding of the compounds to βTrCP was assessed using a fluorescence polarisation (FP) binding assay. The FP assay is an in vitro binding assay using a fluorescein-tagged IkB peptide at 10 nM to mimic substrate binding to βTrCP, and was performed as described above.


Dose response curves for a number of representative peptides are shown in FIGS. 5A-5C.









TABLE 1







DpSGXXpS sequence modified to


determine alternate binding sequences











FP Assay


Entry
Sequence
IC50/μM





 1
D-pS-GIHS-NH2
>100   





 2
GD-pS-GIHS-NH2
>100   





 3
AD-pS-GIHS-NH2
>100   





 4
VD-pS-GIHS-NH2
>100   





 5
DAGIHS-NH2
>100   





 6
GDAGIHS-NH2
>100   





 7
ADAGIHS-NH2
>100   





 8
VDAGIHS-NH2
>100   





 9
DDASGIHS-NH2
>100   





10
LDASGIHS-NH2
>100   





11
LD-pS-SGHIS-NH2
>100   





12
DAGIFE-NH2
>100   





13
EAGIFE-NH2
>100   





14
dEGIFE-NH2
>100   





15
dAGIFE-NH2
>100   





16
dAGIFR-NH2
>100   





17
dNGIFR-NH2
>100   





18
E-pS-GIFE-NH2
  34.5 





19
D-pS-GIFE-NH2
   1.24





20
DAGNFE-NH2
>100   





21
DEGFFE-NH2
  43.6 





22
DAGFFE-NH2
>100   





23
dEGIFD-NH2
>100   





24
dAGIFD-NH2
>100   





25
DAGIFH-NH2
>100   





26
D-pS-GIFH-NH2
>100   





27
D-pS-GNFE-NH2
>100   









The peptide DEGFFE-NH2, having an IC50 of 43.6 μM, was selected as a suitable non-phosphorylated candidate for further progression.


Starting from DEGFFE-NH2, an array of compounds was prepared, as shown in Table 2, in which the N-terminal amino acid (D) was replaced with various capping groups. All of these compounds have an aminde at the C-terminus, as do those shown in Tables 5 and 6. FIG. 2 illustrates the capping groups used and their abbreviations.









TABLE 2







Optimisation of DEGFFE by replacing N-terminal Asp


with capping group





















(SEQ ID



P1
P2
P3
P4
P5
P6
NO:)





 1
D
E
G
I
F
E
(70)


 2
D
E
G
Y
F
E



 3
D
A
G
Y
F
E



 4
Ac
E
G
I
F
E
(71)


 5
MeOCO
E
G
I
F
E



 6
EtOCO
E
G
I
F
E



 7
BnOCO
E
G
I
F
E



 8
4-(MeO)—PhOCO
E
G
I
F
E



 9
4-(NO2)—PhOCO
E
G
I
F
E



10
Piv
E
G
I
F
E



11
Bz
E
G
I
F
E



12
4-(MeO)—PhCO
E
G
I
F
E



13
4-(NO2)—PhCO
E
G
I
F
E



14
Palm
E
G
I
F
E



15
Stear
E
G
I
F
E



16
TPCC
E
G
I
F
E



17
Phth
E
G
I
F
E



18
Suc
E
G
I
F
E



19
PhNHCO
E
G
I
F
E



20
BnNHCO
E
G
I
F
E



21
4-(NO2)—PhNHCO
E
G
I
F
E



22
Ms
E
G
I
F
E



23
PhSO2
E
G
I
F
E



24
Ts
E
G
I
F
E



25
Ns
E
G
I
F
E



26
D
E
G
F
F
E



27
Ac
E
G
F
F
E
(72)


28
MeOCO
E
G
F
F
E



29
EtOCO
E
G
F
F
E



30
BnOCO
E
G
F
F
E



31
4-(MeO)—PhOCO
E
G
F
F
E



32
4-(NO2)—PhOCO
E
G
F
F
E



33
Piv
E
G
F
F
E



34
Bz
E
G
F
F
E



35
4-(MeO)—PhCO
E
G
F
F
E



36
4-(NO2)—PhCO
E
G
F
F
E



37
Palm
E
G
F
F
E



38
Stear
E
G
F
F
E



39
TPCC
E
G
F
F
E



40
Phth
E
G
F
F
E



41
Suc
E
G
F
F
E



42
PhNHCO
E
G
F
F
E



43
BnNHCO
E
G
F
F
E



44
4-(NO2)—PhNHCO
E
G
F
F
E



45
Ms
E
G
F
F
E



46
PhSO2
E
G
F
F
E



47
Ts
E
G
F
F
E



48
Ns
E
G
F
F
E









Four sequences were selected for re-synthesis and testing in the FP assay, which was performed as described above along with 4 negative controls. The results of the FP assay are shown in Table 3.









TABLE 3







FP assay results of selected


capped sequences and negative controls











FP Assay


Entry
Sequence (SEQ ID NO:)
IC50/μM





1
DEGIFE-NH2 (13)
>100   





2
Phth-EGIFE-NH2 (71)
>100   





3
Suc-AGIFE-NH2 (73)
>100   





4
Phth-AGIFE-NH2 (73)
>100   





5
Suc-EGIFE-NH2 (71)
>100   





6
Phth-EGFFE-NH2 (72)
  10.9 





7
Suc-EGFFE-NH2 (57)
   3.18





8
Phth-AGFFE-NH2 (74)
>100   





9
Suc-AGFFE-NH2 (74)
>100   









The FP assay identified Suc-EGFFE-NH2 as a low μM inhibitor for further optimisation.


Further peptides were synthesised as described above by replacing residues in the Suc-EGFFE-NH2 sequence with alternative acidic capping groups and non-natural amino acids. The sequences and abbreviations of the acidic capping groups and non-natural amino acids are shown in FIGS. 3 and 4A-4B, respectively, as well as in Table 4, and the generated peptide sequences are shown in Tables 5 and 6.









TABLE 4





Key to capping groups


















Succinic anhydride
Suc



PEG 2035
Peg35



PEG2030
Peg30



Trans aconitic acid
Taa



Cis aconitic acid
Caa



Fumaric acid
Fum



Terephthalic acid
TA



Isophthalic acid
Ia



1,4 cyclohexanedicarboxylic acid
1,4-Chda



trans-1,2 cyclohexanedicarboxylic acid
1,2-Chda



Glutaric anhydride
Ga

















TABLE 5







Sequence of β-TrCP binding peptides (5-Mers) synthesised















P1
P2
P3
P4
P5
P6
(SEQ ID NO:)





 1
Peg35
E
G
F
F
E
(75)


 2
Peg30
E
G
F
F
E



 3
Taa
E
G
F
F
E



 4
Caa
E
G
F
F
E



 5
Fum
E
G
F
F
E



 6
TA
E
G
F
F
E



 7
Ia
E
G
F
F
E



 8
1,4-Chda
E
G
F
F
E



 9
1,2-Chda
E
G
F
F
E



 10
Ga
E
G
F
F
E



 11
Suc
E
G
Y
F
E



 12
Peg35
E
G
Y
F
E



 13
Peg30
E
G
Y
F
E



 14
Taa
E
G
Y
F
E



 15
Caa
E
G
Y
F
E



 16
Fum
E
G
Y
F
E



 17
TA
E
G
Y
F
E



 18
Ia
E
G
Y
F
E



 19
1,4-Chda
E
G
Y
F
E



 20
1,2-Chda
E
G
Y
F
E



 21
Ga
E
G
Y
F
E



 22
Suc
E
A
F
F
E
(76)


 23
Peg35
E
A
F
F
E



 24
Peg30
E
A
F
F
E



 25
Taa
E
A
F
F
E



 26
Caa
E
A
F
F
E



 27
Fum
E
A
F
F
E



 28
TA
E
A
F
F
E



 29
Ia
E
A
F
F
E



 30
1,4-Chda
E
A
F
F
E



 31
1,2-Chda
E
A
F
F
E



 32
Ga
E
A
F
F
E



 33
Suc
E
A
F
F
E



 34
Peg35
E
A
F
F
E



 35
Peg30
E
A
F
F
E



 36
Taa
E
A
F
F
E



 37
Caa
E
A
F
F
E



 38
Fum
E
A
F
F
E



 39
TA
E
A
F
F
E



 40
Ia
E
A
F
F
E



 41
1,4-Chda
E
A
F
F
E



 42
1,2-Chda
E
A
F
F
E



 43
Ga
E
A
F
F
E



 44
Suc
E
βA
F
F
E
(77)


 45
Peg35
E
βA
F
F
E



 46
Peg30
E
βA
F
F
E



 47
Taa
E
βA
F
F
E



 48
Caa
E
βA
F
F
E



 49
Fum
E
βA
F
F
E



 50
TA
E
βA
F
F
E



 51
Ia
E
βA
F
F
E



 52
1,4-Chda
E
βA
F
F
E



 53
1,2-Chda
E
βA
F
F
E



 54
Ga
E
βA
F
F
E



 55
Suc
E
P
F
F
E
(78)


 56
Peg35
E
P
F
F
E



 57
Peg30
E
P
F
F
E



 58
Taa
E
P
F
F
E



 59
Caa
E
P
F
F
E



 60
Fum
E
P
F
F
E



 61
TA
E
P
F
F
E



 62
Ia
E
P
F
F
E



 63
1,4-Chda
E
P
F
F
E



 64
1,2-Chda
E
P
F
F
E



 65
Ga
E
P
F
F
E



 66
Suc
E
p
F
F
E



 67
Peg35
E
p
F
F
E



 68
Peg30
E
p
F
F
E



 69
Taa
E
p
F
F
E



 70
Caa
E
p
F
F
E



 71
Fum
E
p
F
F
E



 72
TA
E
p
F
F
E



 73
Ia
E
p
F
F
E



 74
1,4-Chda
E
p
F
F
E



 75
1,2-Chda
E
p
F
F
E



 76
Ga
E
p
F
F
E



 77
Suc
E
Sar
F
F
E
(79)


 78
Peg35
E
Sar
F
F
E



 79
Peg30
E
Sar
F
F
E



 80
Taa
E
Sar
F
F
E



 81
Caa
E
Sar
F
F
E



 82
Fum
E
Sar
F
F
E



 83
TA
E
Sar
F
F
E



 84
Ia
E
Sar
F
F
E



 85
1,4-Chda
E
Sar
F
F
E



 86
1,2-Chda
E
Sar
F
F
E



 87
Ga
E
Sar
F
F
E



 88
Suc
E
β-H-ala
F
F
E



 89
Peg35
E
β-H-ala
F
F
E



 90
Peg30
E
β-H-ala
F
F
E



 91
Taa
E
β-H-ala
F
F
E



 92
Caa
E
β-H-ala
F
F
E



 93
Fum
E
β-H-ala
F
F
E



 94
TA
E
β-H-ala
F
F
E



 95
Ia
E
β-H-ala
F
F
E



 96
1,4-Chda
E
β-H-ala
F
F
E



 97
1,2-Chda
E
β-H-ala
F
F
E



 98
Ga
E
β-H-ala
F
F
E



 99
Suc
E
G
F
F
E
(80)


100
Suc
Q
G
F
F
E



101
Suc
N
G
F
F
E



102
Suc
(Me)E
G
F
F
E



103
Suc
βE
G
F
F
E



104
Suc
(Me)D
G
F
F
E



105
Suc
βD
G
F
F
E



106
Suc
E(Me)
G
F
F
E



107
Suc
D
G
F
F
E



108
Suc
Gla
G
F
F
E



109
Suc
E
G
Y
F
E



110
Suc
Q
G
Y
F
E



111
Suc
N
G
Y
F
E



112
Suc
(Me)E
G
Y
F
E



113
Suc
βE
G
Y
F
E



114
Suc
(Me)D
G
Y
F
E



115
Suc
βD
G
Y
F
E



116
Suc
E(Me)
G
Y
F
E



117
Suc
D
G
Y
F
E



118
Suc
Gla
G
Y
F
E



119
Suc
E
G
W
F
E



120
Suc
Q
G
W
F
E



121
Suc
N
G
W
F
E



122
Suc
(Me)E
G
W
F
E



123
Suc
βE
G
W
F
E



124
Suc
(Me)D
G
W
F
E



125
Suc
βD
G
W
F
E



126
Suc
E(Me)
G
W
F
E



127
Suc
D
G
W
F
E



128
Suc
Gla
G
W
F
E



129
Suc
E
G
F(3Br)
F
E
(81)


130
Suc
Q
G
F(3Br)
F
E



131
Suc
N
G
F(3Br)
F
E



132
Suc
(Me)E
G
F(3Br)
F
E



133
Suc
βE
G
F(3Br)
F
E



134
Suc
(Me)D
G
F(3Br)
F
E



135
Suc
βD
G
F(3Br)
F
E



136
Suc
E(Me)
G
F(3Br)
F
E



137
Suc
D
G
F(3Br)
F
E



138
Suc
Gla
G
F(3Br)
F
E



139
Suc
E
G
F(4NO2)
F
E



140
Suc
Q
G
F(4NO2)
F
E



141
Suc
N
G
F(4NO2)
F
E



142
Suc
(Me)E
G
F(4NO2)
F
E



143
Suc
βE
G
F(4NO2)
F
E



144
Suc
(Me)D
G
F(4NO2)
F
E



145
Suc
βD
G
F(4NO2)
F
E



146
Suc
E(Me)
G
F(4NO2)
F
E



147
Suc
D
G
F(4NO2)
F
E



148
Suc
Gla
G
F(4NO2)
F
E



149
Suc
E
G
3Pal
F
E
(82)


150
Suc
Q
G
3Pal
F
E



151
Suc
N
G
3Pal
F
E



152
Suc
(Me)E
G
3Pal
F
E



153
Suc
βE
G
3Pal
F
E



154
Suc
(Me)D
G
3Pal
F
E



155
Suc
βD
G
3Pal
F
E



156
Suc
E(Me)
G
3Pal
F
E



157
Suc
D
G
3Pal
F
E



158
Suc
Gla
G
3Pal
F
E



159
Suc
E
G
Y(Me)
F
E



160
Suc
Q
G
Y(Me)
F
E



161
Suc
N
G
Y(Me)
F
E



162
Suc
(Me)E
G
Y(Me)
F
E



163
Suc
βE
G
Y(Me)
F
E



164
Suc
(Me)D
G
Y(Me)
F
E



165
Suc
βD
G
Y(Me)
F
E



166
Suc
E(Me)
G
Y(Me)
F
E



167
Suc
D
G
Y(Me)
F
E



168
Suc
Gla
G
Y(Me)
F
E



169
Suc
E
G
Pip
F
E



170
Suc
Q
G
Pip
F
E



171
Suc
N
G
Pip
F
E



172
Suc
(Me)E
G
Pip
F
E



173
Suc
βE
G
Pip
F
E



174
Suc
(Me)D
G
Pip
F
E



175
Suc
βD
G
Pip
F
E



176
Suc
E(Me)
G
Pip
F
E



177
Suc
D
G
Pip
F
E



178
Suc
Gla
G
Pip
F
E



179
Suc
E
G
Tic
F
E



180
Suc
Q
G
Tic
F
E



181
Suc
N
G
Tic
F
E



182
Suc
(Me)E
G
Tic
F
E



183
Suc
βE
G
Tic
F
E



184
Suc
(Me)D
G
Tic
F
E



185
Suc
βD
G
Tic
F
E



186
Suc
E(Me)
G
Tic
F
E



187
Suc
D
G
Tic
F
E



188
Suc
Gla
G
Tic
F
E



189
Suc
E
G
1Nal
F
E



190
Suc
Q
G
1Nal
F
E



191
Suc
N
G
1Nal
F
E



192
Suc
(Me)E
G
1Nal
F
E



193
Suc
βE
G
1Nal
F
E



194
Suc
(Me)D
G
1Nal
F
E



195
Suc
βD
G
1Nal
F
E



196
Suc
E(Me)
G
1Nal
F
E



197
Suc
D
G
1Nal
F
E



198
Suc
Gla
G
1Nal
F
E



199
Suc
E
G
F(2F)
F
E
(83)


200
Suc
Q
G
F(2F)
F
E



201
Suc
N
G
F(2F)
F
E



202
Suc
(Me)E
G
F(2F)
F
E



203
Suc
βE
G
F(2F)
F
E



204
Suc
(Me)D
G
F(2F)
F
E



205
Suc
βD
G
F(2F)
F
E



206
Suc
E(Me)
G
F(2F)
F
E



207
Suc
D
G
F(2F)
F
E



208
Suc
Gla
G
F(2F)
F
E



209
Suc
E
G
F(3F)
F
E



210
Suc
Q
G
F(3F)
F
E



211
Suc
N
G
F(3F)
F
E



212
Suc
(Me)E
G
F(3F)
F
E



213
Suc
βE
G
F(3F)
F
E



214
Suc
(Me)D
G
F(3F)
F
E



215
Suc
βD
G
F(3F)
F
E



216
Suc
E(Me)
G
F(3F)
F
E



217
Suc
D
G
F(3F)
F
E



218
Suc
Gla
G
F(3F)
F
E



219
Suc
E
G
F(4F)
F
E



220
Suc
Q
G
F(4F)
F
E



221
Suc
N
G
F(4F)
F
E



222
Suc
(Me)E
G
F(4F)
F
E



223
Suc
βE
G
F(4F)
F
E



224
Suc
(Me)D
G
F(4F)
F
E



225
Suc
βD
G
F(4F)
F
E



226
Suc
E(Me)
G
F(4F)
F
E



227
Suc
D
G
F(4F)
F
E



228
Suc
Gla
G
F(4F)
F
E



229
Suc
E
G
F
F
E
(84)


230
Suc
E
G
F
F
Q



231
Suc
E
G
F
F
N



232
Suc
E
G
F
F
(Me)E



233
Suc
E
G
F
F
βE



234
Suc
E
G
F
F
(Me)D



235
Suc
E
G
F
F
βD



236
Suc
E
G
F
F
E(Me)



237
Suc
E
G
F
F
D



238
Suc
E
G
F
F
Gla



239
Suc
E
G
F
Y
E



240
Suc
E
G
F
Y
Q



241
Suc
E
G
F
Y
N



242
Suc
E
G
F
Y
(Me)E



243
Suc
E
G
F
Y
βE



244
Suc
E
G
F
Y
(Me)D



245
Suc
E
G
F
Y
βD



246
Suc
E
G
F
Y
E(Me)



247
Suc
E
G
F
Y
D



248
Suc
E
G
F
Y
Gla



249
Suc
E
G
F
W
E



250
Suc
E
G
F
W
Q



251
Suc
E
G
F
W
N



252
Suc
E
G
F
W
(Me)E



253
Suc
E
G
F
W
βE



254
Suc
E
G
F
W
(Me)D



255
Suc
E
G
F
W
βD



256
Suc
E
G
F
W
E(Me)



257
Suc
E
G
F
W
D



258
Suc
E
G
F
W
Gla



259
Suc
E
G
F
F(3Br)
E
(85)


260
Suc
E
G
F
F(3Br)
Q



261
Suc
E
G
F
F(3Br)
N



262
Suc
E
G
F
F(3Br)
(Me)E



263
Suc
E
G
F
F(3Br)
βE



264
Suc
E
G
F
F(3Br)
(Me)D



265
Suc
E
G
F
F(3Br)
βD



266
Suc
E
G
F
F(3Br)
E(Me)



267
Suc
E
G
F
F(3Br)
D



268
Suc
E
G
F
F(3Br)
Gla



269
Suc
E
G
F
F(4NO2)
E



270
Suc
E
G
F
F(4NO2)
Q



271
Suc
E
G
F
F(4NO2)
N



272
Suc
E
G
F
F(4NO2)
(Me)E



273
Suc
E
G
F
F(4NO2)
r3E



274
Suc
E
G
F
F(4NO2)
(Me)D



275
Suc
E
G
F
F(4NO2)
βD



276
Suc
E
G
F
F(4NO2)
E(Me)



277
Suc
E
G
F
F(4NO2)
D



278
Suc
E
G
F
F(4NO2)
Gla



279
Suc
E
G
F
3Pal
E
(86)


280
Suc
E
G
F
3Pal
Q



281
Suc
E
G
F
3Pal
N



282
Suc
E
G
F
3Pal
(Me)E



283
Suc
E
G
F
3Pal
βE



284
Suc
E
G
F
3Pal
(Me)D



285
Suc
E
G
F
3Pal
βD



286
Suc
E
G
F
3Pal
E(Me)



287
Suc
E
G
F
3Pal
D



288
Suc
E
G
F
3Pal
Gla



289
Suc
E
G
F
Y(Me)
E



290
Suc
E
G
F
Y(Me)
Q



291
Suc
E
G
F
Y(Me)
N



292
Suc
E
G
F
Y(Me)
(Me)E



293
Suc
E
G
F
Y(Me)
βE



294
Suc
E
G
F
Y(Me)
(Me)D



295
Suc
E
G
F
Y(Me)
βD



296
Suc
E
G
F
Y(Me)
E(Me)



297
Suc
E
G
F
Y(Me)
D



298
Suc
E
G
F
Y(Me)
Gla



299
Suc
E
G
F
Pip
E



300
Suc
E
G
F
Pip
Q



301
Suc
E
G
F
Pip
N



302
Suc
E
G
F
Pip
(Me)E



303
Suc
E
G
F
Pip
βE



304
Suc
E
G
F
Pip
(Me)D



305
Suc
E
G
F
Pip
βD



306
Suc
E
G
F
Pip
E(Me)



307
Suc
E
G
F
Pip
D



308
Suc
E
G
F
Pip
Gla



309
Suc
E
G
F
Tic
E



310
Suc
E
G
F
Tic
Q



311
Suc
E
G
F
Tic
N



312
Suc
E
G
F
Tic
(Me)E



313
Suc
E
G
F
Tic
βE



314
Suc
E
G
F
Tic
(Me)D



315
Suc
E
G
F
Tic
βD



316
Suc
E
G
F
Tic
E(Me)



317
Suc
E
G
F
Tic
D



318
Suc
E
G
F
Tic
Gla



319
Suc
E
G
F
1Nal
E



320
Suc
E
G
F
1Nal
Q



321
Suc
E
G
F
1Nal
N



322
Suc
E
G
F
1Nal
(Me)E



323
Suc
E
G
F
1Nal
βE



324
Suc
E
G
F
1Nal
(Me)D



325
Suc
E
G
F
1Nal
βD



326
Suc
E
G
F
1Nal
E(Me)



327
Suc
E
G
F
1Nal
D



328
Suc
E
G
F
1Nal
Gla



329
Suc
E
G
F
F(2F)
E
(87)


330
Suc
E
G
F
F(2F)
Q



331
Suc
E
G
F
F(2F)
N



332
Suc
E
G
F
F(2F)
(Me)E



333
Suc
E
G
F
F(2F)
βE



334
Suc
E
G
F
F(2F)
(Me)D



335
Suc
E
G
F
F(2F)
βD



336
Suc
E
G
F
F(2F)
E(Me)



337
Suc
E
G
F
F(2F)
D



338
Suc
E
G
F
F(2F)
Gla



339
Suc
E
G
F
F(3F)
E



340
Suc
E
G
F
F(3F)
Q



341
Suc
E
G
F
F(3F)
N



342
Suc
E
G
F
F(3F)
(Me)E



343
Suc
E
G
F
F(3F)
βE



344
Suc
E
G
F
F(3F)
(Me)D



345
Suc
E
G
F
F(3F)
βD



346
Suc
E
G
F
F(3F)
E(Me)



347
Suc
E
G
F
F(3F)
D



348
Suc
E
G
F
F(3F)
Gla



349
Suc
E
G
F
F(4F)
E



350
Suc
E
G
F
F(4F)
Q



351
Suc
E
G
F
F(4F)
N



352
Suc
E
G
F
F(4F)
(Me)E



353
Suc
E
G
F
F(4F)
βE



354
Suc
E
G
F
F(4F)
(Me)D



355
Suc
E
G
F
F(4F)
βD



356
Suc
E
G
F
F(4F)
E(Me)



357
Suc
E
G
F
F(4F)
D



358
Suc
E
G
F
F(4F)
Gla



359
Suc
E
G
F
F
E
(88)


360
Suc
E
G
Y
F
E



361
Suc
E
G
W
F
E



362
Suc
E
G
F(3Br)
F
E



363
Suc
E
G
F(4NO2)
F
E



364
Suc
E
G
3Pal
F
E



365
Suc
E
G
Y(Me)
F
E



366
Suc
E
G
Pip
F
E



367
Suc
E
G
Tic
F
E



368
Suc
E
G
1Nal
F
E



369
Suc
E
G
F(2F)
F
E



370
Suc
E
G
F(3F)
F
E



371
Suc
E
G
F(4F)
F
E



372
Suc
E
G
F
Y
E



373
Suc
E
G
Y
Y
E



374
Suc
E
G
W
Y
E



375
Suc
E
G
F(3Br)
Y
E



376
Suc
E
G
F(4NO2)
Y
E



377
Suc
E
G
3Pal
Y
E



378
Suc
E
G
Y(Me)
Y
E



379
Suc
E
G
Pip
Y
E



380
Suc
E
G
Tic
Y
E



381
Suc
E
G
1Nal
Y
E



382
Suc
E
G
F(2F)
Y
E



383
Suc
E
G
F(3F)
Y
E



384
Suc
E
G
F(4F)
Y
E



385
Suc
E
G
F
W
E



386
Suc
E
G
Y
W
E



387
Suc
E
G
W
W
E



388
Suc
E
G
F(3Br)
W
E



389
Suc
E
G
F(4NO2)
W
E



390
Suc
E
G
3Pal
W
E



391
Suc
E
G
Y(Me)
W
E



392
Suc
E
G
Pip
W
E



393
Suc
E
G
Tic
W
E



394
Suc
E
G
1Nal
W
E



395
Suc
E
G
F(2F)
W
E



396
Suc
E
G
F(3F)
W
E



397
Suc
E
G
F(4F)
W
E



398
Suc
E
G
F
F(3Br)
E
(89)


399
Suc
E
G
Y
F(3Br)
E



400
Suc
E
G
W
F(3Br)
E



401
Suc
E
G
F(3Br)
F(3Br)
E



402
Suc
E
G
F(4NO2)
F(3Br)
E



403
Suc
E
G
3Pal
F(3Br)
E



404
Suc
E
G
Y(Me)
F(3Br)
E



405
Suc
E
G
Pip
F(3Br)
E



406
Suc
E
G
Tic
F(3Br)
E



407
Suc
E
G
1Nal
F(3Br)
E



408
Suc
E
G
F(2F)
F(3Br)
E



409
Suc
E
G
F(3F)
F(3Br)
E



410
Suc
E
G
F(4F)
F(3Br)
E



411
Suc
E
G
F
F(4NO2)
E



412
Suc
E
G
Y
F(4NO2)
E



413
Suc
E
G
W
F(4NO2)
E



414
Suc
E
G
F(3Br)
F(4NO2)
E



415
Suc
E
G
F(4NO2)
F(4NO2)
E



416
Suc
E
G
3Pal
F(4NO2)
E



417
Suc
E
G
Y(Me)
F(4NO2)
E



418
Suc
E
G
Pip
F(4NO2)
E



419
Suc
E
G
Tic
F(4NO2)
E



420
Suc
E
G
1Nal
F(4NO2)
E



421
Suc
E
G
F(2F)
F(4NO2)
E



422
Suc
E
G
F(3F)
F(4NO2)
E



423
Suc
E
G
F(4F)
F(4NO2)
E



424
Suc
E
G
F
3Pal
E



425
Suc
E
G
Y
3Pal
E



426
Suc
E
G
W
3Pal
E



427
Suc
E
G
F(3Br)
3Pal
E



428
Suc
E
G
F(4NO2)
3Pal
E



429
Suc
E
G
3Pal
3Pal
E



430
Suc
E
G
Y(Me)
3Pal
E



431
Suc
E
G
Pip
3Pal
E



432
Suc
E
G
Tic
3Pal
E



433
Suc
E
G
1Nal
3Pal
E



434
Suc
E
G
F(2F)
3Pal
E



435
Suc
E
G
F(3F)
3Pal
E



436
Suc
E
G
F(4F)
3Pal
E



437
Suc
E
G
F
Y(Me)
E



438
Suc
E
G
Y
Y(Me)
E



439
Suc
E
G
W
Y(Me)
E



440
Suc
E
G
F(3Br)
Y(Me)
E



441
Suc
E
G
F(4NO2)
Y(Me)
E



442
Suc
E
G
3Pal
Y(Me)
E



443
Suc
E
G
Y(Me)
Y(Me)
E



444
Suc
E
G
Pip
Y(Me)
E



445
Suc
E
G
Tic
Y(Me)
E



446
Suc
E
G
1Nal
Y(Me)
E



447
Suc
E
G
F(2F)
Y(Me)
E



448
Suc
E
G
F(3F)
Y(Me)
E



449
Suc
E
G
F(4F)
Y(Me)
E



450
Suc
E
G
F
Pip
E



451
Suc
E
G
Y
Pip
E



452
Suc
E
G
W
Pip
E



453
Suc
E
G
F(3Br)
Pip
E



454
Suc
E
G
F(4NO2)
Pip
E



455
Suc
E
G
3Pal
Pip
E



456
Suc
E
G
Y(Me)
Pip
E



457
Suc
E
G
Pip
Pip
E



458
Suc
E
G
Tic
Pip
E



459
Suc
E
G
1Nal
Pip
E



460
Suc
E
G
F(2F)
Pip
E



461
Suc
E
G
F(3F)
Pip
E



462
Suc
E
G
F(4F)
Pip
E



463
Suc
E
G
F
Tic
E



464
Suc
E
G
Y
Tic
E



465
Suc
E
G
W
Tic
E



466
Suc
E
G
F(3Br)
Tic
E



467
Suc
E
G
F(4NO2)
Tic
E



468
Suc
E
G
3Pal
Tic
E



469
Suc
E
G
Y(Me)
Tic
E



470
Suc
E
G
Pip
Tic
E



471
Suc
E
G
Tic
Tic
E



472
Suc
E
G
1Nal
Tic
E



473
Suc
E
G
F(2F)
Tic
E



474
Suc
E
G
F(3F)
Tic
E



475
Suc
E
G
F(4F)
Tic
E



476
Suc
E
G
F
1Nal
E



477
Suc
E
G
Y
1Nal
E



478
Suc
E
G
W
1Nal
E



479
Suc
E
G
F(3Br)
1Nal
E



480
Suc
E
G
F(4NO2)
1Nal
E



481
Suc
E
G
3Pal
1Nal
E



482
Suc
E
G
Y(Me)
1Nal
E



483
Suc
E
G
Pip
1Nal
E



484
Suc
E
G
Tic
1Nal
E



485
Suc
E
G
1Nal
1Nal
E



486
Suc
E
G
F(2F)
1Nal
E



487
Suc
E
G
F(3F)
1Nal
E



488
Suc
E
G
F(4F)
1Nal
E



489
Suc
E
G
F
F(2F)
E



490
Suc
E
G
Y
F(2F)
E



491
Suc
E
G
W
F(2F)
E



492
Suc
E
G
F(3Br)
F(2F)
E



493
Suc
E
G
F(4NO2)
F(2F)
E



494
Suc
E
G
3Pal
F(2F)
E



495
Suc
E
G
Y(Me)
F(2F)
E



496
Suc
E
G
Pip
F(2F)
E



497
Suc
E
G
Tic
F(2F)
E



498
Suc
E
G
1Nal
F(2F)
E



499
Suc
E
G
F(2F)
F(2F)
E



500
Suc
E
G
F(3F)
F(2F)
E



501
Suc
E
G
F(4F)
F(2F)
E



502
Suc
E
G
F
F(3F)
E



503
Suc
E
G
Y
F(3F)
E



504
Suc
E
G
W
F(3F)
E



505
Suc
E
G
F(3Br)
F(3F)
E



506
Suc
E
G
F(4NO2)
F(3F)
E



507
Suc
E
G
3Pal
F(3F)
E



508
Suc
E
G
Y(Me)
F(3F)
E



509
Suc
E
G
Pip
F(3F)
E



510
Suc
E
G
Tic
F(3F)
E



511
Suc
E
G
1Nal
F(3F)
E



512
Suc
E
G
F(2F)
F(3F)
E



513
Suc
E
G
F(3F)
F(3F)
E



514
Suc
E
G
F(4F)
F(3F)
E



515
Suc
E
G
F
F(4F)
E



516
Suc
E
G
Y
F(4F)
E



517
Suc
E
G
W
F(4F)
E



518
Suc
E
G
F(3Br)
F(4F)
E



519
Suc
E
G
F(4NO2)
F(4F)
E



520
Suc
E
G
3Pal
F(4F)
E



521
Suc
E
G
Y(Me)
F(4F)
E



522
Suc
E
G
Pip
F(4F)
E



523
Suc
E
G
Tic
F(4F)
E



524
Suc
E
G
1Nal
F (4F)
E



525
Suc
E
G
F (2F)
F (4F)
E



526
Suc
E
G
F (3F)
F (4F)
E



527
Suc
E
G
F (4F)
F (4F)
E



528
Suc
E
G
F
F
E
(90)


529
Suc
Q
G
F
F
E



530
Suc
N
G
F
F
E



531
Suc
(Me)E
G
F
F
E



532
Suc
βE
G
F
F
E



533
Suc
(Me)D
G
F
F
E



534
Suc
βD
G
F
F
E



535
Suc
E(Me)
G
F
F
E



536
Suc
D
G
F
F
E



537
Suc
Gla
G
F
F
E



538
Suc
Q
G
F
F
Q



539
Suc
N
G
F
F
Q



540
Suc
(Me)E
G
F
F
Q



541
Suc
βE
G
F
F
Q



542
Suc
(Me)D
G
F
F
Q



543
Suc
βD
G
F
F
Q



544
Suc
E(Me)
G
F
F
Q



545
Suc
D
G
F
F
Q



546
Suc
Gla
G
F
F
Q



547
Suc
E
G
F
F
Q



548
Suc
Q
G
F
F
N



549
Suc
N
G
F
F
N



550
Suc
(Me)E
G
F
F
N



551
Suc
βE
G
F
F
N



552
Suc
(Me)D
G
F
F
N



553
Suc
βD
G
F
F
N



554
Suc
E(Me)
G
F
F
N



555
Suc
D
G
F
F
N



556
Suc
Gla
G
F
F
N



557
Suc
E
G
F
F
N



558
Suc
Q
G
F
F
(Me)E



559
Suc
N
G
F
F
(Me)E



560
Suc
(Me)E
G
F
F
(Me)E



561
Suc
βE
G
F
F
(Me)E



562
Suc
(Me)D
G
F
F
(Me)E



563
Suc
βD
G
F
F
(Me)E



564
Suc
E(Me)
G
F
F
(Me)E



565
Suc
D
G
F
F
(Me)E



566
Suc
Gla
G
F
F
(Me)E



567
Suc
E
G
F
F
(Me)E



568
Suc
Q
G
F
F
βE



569
Suc
N
G
F
F
βE



570
Suc
(Me)E
G
F
F
βE



571
Suc
βE
G
F
F
βE



572
Suc
(Me)D
G
F
F
βE



573
Suc
βD
G
F
F
βE



574
Suc
E(Me)
G
F
F
βE



575
Suc
D
G
F
F
βE



576
Suc
Gla
G
F
F
βE



577
Suc
E
G
F
F
βE



578
Suc
Q
G
F
F
(Me)D



579
Suc
N
G
F
F
(Me)D



580
Suc
(Me)E
G
F
F
(Me)D



581
Suc
βE
G
F
F
(Me)D



582
Suc
(Me)D
G
F
F
(Me)D



583
Suc
βD
G
F
F
(Me)D



584
Suc
E(Me)
G
F
F
(Me)D



585
Suc
D
G
F
F
(Me)D



586
Suc
Gla
G
F
F
(Me)D



587
Suc
E
G
F
F
(Me)D



588
Suc
Q
G
F
F
βD



589
Suc
N
G
F
F
βD



590
Suc
(Me)E
G
F
F
βD



591
Suc
βE
G
F
F
βD



592
Suc
(Me)D
G
F
F
βD



593
Suc
βD
G
F
F
βD



594
Suc
E(Me)
G
F
F
βD



595
Suc
D
G
F
F
βD



596
Suc
Gla
G
F
F
βD



597
Suc
E
G
F
F
βD



598
Suc
Q
G
F
F
E(Me)



599
Suc
N
G
F
F
E(Me)



600
Suc
(Me)E
G
F
F
E(Me)



601
Suc
βE
G
F
F
E(Me)



602
Suc
(Me)D
G
F
F
E(Me)



603
Suc
βD
G
F
F
E(Me)



604
Suc
E(Me)
G
F
F
E(Me)



605
Suc
D
G
F
F
E(Me)



606
Suc
Gla
G
F
F
E(Me)



607
Suc
E
G
F
F
E(Me)



608
Suc
Q
G
F
F
D



609
Suc
N
G
F
F
D



610
Suc
(Me)E
G
F
F
D



611
Suc
βE
G
F
F
D



612
Suc
(Me)D
G
F
F
D



613
Suc
βD
G
F
F
D



614
Suc
E(Me)
G
F
F
D



615
Suc
D
G
F
F
D



616
Suc
Gla
G
F
F
D



617
Suc
E
G
F
F
D



618
Suc
Q
G
F
F
Gla



619
Suc
N
G
F
F
Gla



620
Suc
(Me)E
G
F
F
Gla



621
Suc
βE
G
F
F
Gla



622
Suc
(Me)D
G
F
F
Gla



623
Suc
βD
G
F
F
Gla



624
Suc
E(Me)
G
F
F
Gla



625
Suc
D
G
F
F
Gla



626
Suc
Gla
G
F
F
Gla



627
Suc
E
G
F
F
Gla
(91)


628
Suc
E
G
F
F
E



629
Suc
E
G
F
F
F



630
Suc
E
G
F
F
Y



631
Suc
E
G
F
F
W



632
Suc
E
G
F
F
F(3Br)



633
Suc
E
G
F
F
F(4NO2)



634
Suc
E
G
F
F
3Pal



635
Suc
E
G
F
F
Y(Me)



636
Suc
E
G
F
F
Pip



637
Suc
E
G
F
F
Tic



638
Suc
E
G
F
F
1Nal



639
Suc
E
G
F
F
F(2F)



640
Suc
E
G
F
F
F(3F)



641
Suc
E
G
F
F
F(4F)



642
Suc
E
G
Y
F
E



643
Suc
E
G
Y
F
F



644
Suc
E
G
Y
F
Y



645
Suc
E
G
Y
F
W



646
Suc
E
G
Y
F
F(3Br)



647
Suc
E
G
Y
F
F(4NO2)



648
Suc
E
G
Y
F
3Pal



649
Suc
E
G
Y
F
Y(Me)



650
Suc
E
G
Y
F
Pip



651
Suc
E
G
Y
F
Tic



652
Suc
E
G
Y
F
1Nal



653
Suc
E
G
Y
F
F(2F)



654
Suc
E
G
Y
F
F(3F)



655
Suc
E
G
Y
F
F(4F)



656
Suc
E
G
F
F
E
(92)


657
Suc
E
A
F
F
E



658
Suc
E
a
F
F
E



659
Suc
E
βA
F
F
E



660
Suc
E
P
F
F
E



661
Suc
E
p
F
F
E



662
Suc
E
Sar
F
F
E



663
Suc
E
β-H-ala
F
F
E



664
Suc
E
G
Y
F
E



665
Suc
E
A
Y
F
E



666
Suc
E
a
Y
F
E



667
Suc
E
βA
Y
F
E



668
Suc
E
P
Y
F
E



669
Suc
E
p
Y
F
E



670
Suc
E
Sar
Y
F
E



671
Suc
E
β-H-ala
Y
F
E



672
Suc
E
G
F
F
E
(93)


673
Suc
E
G
F
F
E(Me)



674
Suc
E
G
Y
F
E(Me)



675
Suc
E
G
W
F
E(Me)



676
Suc
E
G
1Nal
F
E(Me)



677
Suc
E
G
Tic
F
E(Me)



678
Suc
E
G
Pip
F
E(Me)



679
Suc
E
G
F(3Br)
F
E(Me)



680
Suc
E
G
3Pal
F
E(Me)



681
Suc
E
G
F
Y
E(Me)



682
Suc
E
G
Y
Y
E(Me)



683
Suc
E
G
W
Y
E(Me)



684
Suc
E
G
1Nal
Y
E(Me)



685
Suc
E
G
Tic
Y
E(Me)



686
Suc
E
G
Pip
Y
E(Me)



687
Suc
E
G
F(3Br)
Y
E(Me)



688
Suc
E
G
3Pal
Y
E(Me)



689
Suc
E
G
F
W
E(Me)



690
Suc
E
G
Y
W
E(Me)



691
Suc
E
G
W
W
E(Me)



692
Suc
E
G
1Nal
W
E(Me)



693
Suc
E
G
Tic
W
E(Me)



694
Suc
E
G
Pip
W
E(Me)



695
Suc
E
G
F(3Br)
W
E(Me)



696
Suc
E
G
3Pal
W
E(Me)



697
Suc
E
G
F
1Nal
E(Me)



698
Suc
E
G
Y
1Nal
E(Me)



699
Suc
E
G
W
1Nal
E(Me)



700
Suc
E
G
1Nal
1Nal
E(Me)



701
Suc
E
G
Tic
1Nal
E(Me)



702
Suc
E
G
Pip
1Nal
E(Me)



703
Suc
E
G
F(3Br)
1Nal
E(Me)



704
Suc
E
G
3Pal
1Nal
E(Me)



705
Suc
E
G
F
Tic
E(Me)



706
Suc
E
G
Y
Tic
E(Me)



707
Suc
E
G
W
Tic
E(Me)



708
Suc
E
G
1Nal
Tic
E(Me)



709
Suc
E
G
Tic
Tic
E(Me)



710
Suc
E
G
Pip
Tic
E(Me)



711
Suc
E
G
F(3Br)
Tic
E(Me)



712
Suc
E
G
3Pal
Tic
E(Me)



713
Suc
E
G
F
Pip
E(Me)



714
Suc
E
G
Y
Pip
E(Me)



715
Suc
E
G
W
Pip
E(Me)



716
Suc
E
G
1Nal
Pip
E(Me)



717
Suc
E
G
Tic
Pip
E(Me)



718
Suc
E
G
Pip
Pip
E(Me)



719
Suc
E
G
F(3Br)
Pip
E(Me)



720
Suc
E
G
3Pal
Pip
E(Me)



721
Suc
E
G
F
F(3Br)
E(Me)



722
Suc
E
G
Y
F(3Br)
E(Me)



723
Suc
E
G
W
F(3Br)
E(Me)



724
Suc
E
G
1Nal
F(3Br)
E(Me)



725
Suc
E
G
Tic
F(3Br)
E(Me)



726
Suc
E
G
Pip
F(3Br)
E(Me)



727
Suc
E
G
F(3Br)
F(3Br)
E(Me)



728
Suc
E
G
3Pal
F(3Br)
E(Me)



729
Suc
E
G
F
3Pal
E(Me)



730
Suc
E
G
Y
3Pal
E(Me)



731
Suc
E
G
W
3Pal
E(Me)



732
Suc
E
G
1Nal
3Pal
E(Me)



733
Suc
E
G
Tic
3Pal
E(Me)



734
Suc
E
G
Pip
3Pal
E(Me)



735
Suc
E
G
F(3Br)
3Pal
E(Me)



736
Suc
E
G
3Pal
3Pal
E(Me)



737
Suc
E
Ahx
F
F
E
(94)


738
Suc
Ahx
G
F
F
E



739
Suc
E
Ahx
F
F
E



740
Suc
E
G
Ahx
F
E



741
Suc
E
G
F
Ahx
E
















TABLE 6







Sequence of (βTrCP binding peptides (4-mers) synthesised



















(SEQ ID



P1
P2
P3
P4
P5
NO:)





 1
Suc
E
G
F
F
 (95)


 2
Suc
E
G
Y
F



 3
Suc
E
G
W
F



 4
Suc
E
G
F(3Br)
F



 5
Suc
E
G
F(4NO2)
F



 6
Suc
E
G
3Pal
F



 7
Suc
E
G
Y(Me)
F



 8
Suc
E
G
Pip
F



 9
Suc
E
G
Tic
F



 10
Suc
E
G
1Nal
F



 11
Suc
E
G
F(2F)
F



 12
Suc
E
G
F(3F)
F



 13
Suc
E
G
F(4F)
F



 14
Suc
E
G
F
Y



 15
Suc
E
G
Y
Y



 16
Suc
E
G
W
Y



 17
Suc
E
G
F(3Br)
Y



 18
Suc
E
G
F(4NO2)
Y



 19
Suc
E
G
3Pal
Y



 20
Suc
E
G
Y(Me)
Y



 21
Suc
E
G
Pip
Y



 22
Suc
E
G
Tic
Y



 23
Suc
E
G
1Nal
Y



 24
Suc
E
G
F(2F)
Y



 25
Suc
E
G
F(3F)
Y



 26
Suc
E
G
F(4F)
Y



 27
Suc
E
G
F
W



 28
Suc
E
G
Y
W



 29
Suc
E
G
W
W



 30
Suc
E
G
F(3Br)
W



 31
Suc
E
G
F(4NO2)
W



 32
Suc
E
G
3Pal
W



 33
Suc
E
G
Y(Me)
W



 34
Suc
E
G
Pip
W



 35
Suc
E
G
Tic
W



 36
Suc
E
G
1Nal
W



 37
Suc
E
G
F(2F)
W



 38
Suc
E
G
F(3F)
W



 39
Suc
E
G
F(4F)
W



 40
Suc
E
G
F
F(3Br)



 41
Suc
E
G
Y
F(3Br)



 42
Suc
E
G
W
F(3Br)



 43
Suc
E
G
F(3Br)
F(3Br)



 44
Suc
E
G
F(4NO2)
F(3Br)



 45
Suc
E
G
3Pal
F(3Br)



 46
Suc
E
G
Y(Me)
F(3Br)



 47
Suc
E
G
Pip
F(3Br)



 48
Suc
E
G
Tic
F(3Br)



 49
Suc
E
G
1Nal
F(3Br)



 50
Suc
E
G
F(2F)
F(3Br)



 51
Suc
E
G
F(3F)
F(3Br)



 52
Suc
E
G
F(4F)
F(3Br)



 53
Suc
E
G
F
F(4NO2)



 54
Suc
E
G
Y
F(4NO2)



 55
Suc
E
G
W
F(4NO2)



 56
Suc
E
G
F(3Br)
F(4NO2)



 57
Suc
E
G
F(4NO2)
F(4NO2)



 58
Suc
E
G
3Pal
F(4NO2)



 59
Suc
E
G
Y(Me)
F(4NO2)



 60
Suc
E
G
Pip
F(4NO2)



 61
Suc
E
G
Tic
F(4NO2)



 62
Suc
E
G
1Nal
F(4NO2)



 63
Suc
E
G
F(2F)
F(4NO2)



 64
Suc
E
G
F(3F)
F(4NO2)



 65
Suc
E
G
F(4F)
F(4NO2)



 66
Suc
E
G
F
3Pal



 67
Suc
E
G
Y
3Pal



 68
Suc
E
G
W
3Pal



 69
Suc
E
G
F(3Br)
3Pal



 70
Suc
E
G
F(4NO2)
3Pal



 71
Suc
E
G
3Pal
3Pal



 72
Suc
E
G
Y(Me)
3Pal



 73
Suc
E
G
Pip
3Pal



 74
Suc
E
G
Tic
3Pal



 75
Suc
E
G
1Nal
3Pal



 76
Suc
E
G
F(2F)
3Pal



 77
Suc
E
G
F(3F)
3Pal



 78
Suc
E
G
F(4F)
3Pal



 79
Suc
E
G
F
Y(Me)



 80
Suc
E
G
Y
Y(Me)



 81
Suc
E
G
W
Y(Me)



 82
Suc
E
G
F(3Br)
Y(Me)



 83
Suc
E
G
F(4NO2)
Y(Me)



 84
Suc
E
G
3Pal
Y(Me)



 85
Suc
E
G
Y(Me)
Y(Me)



 86
Suc
E
G
Pip
Y(Me)



 87
Suc
E
G
Tic
Y(Me)



 88
Suc
E
G
1Nal
Y(Me)



 89
Suc
E
G
F(2F)
Y(Me)



 90
Suc
E
G
F(3F)
Y(Me)



 91
Suc
E
G
F(4F)
Y(Me)



 92
Suc
E
G
F
Pip



 93
Suc
E
G
Y
Pip



 94
Suc
E
G
W
Pip



 95
Suc
E
G
F(3Br)
Pip



 96
Suc
E
G
F(4NO2)
Pip



 97
Suc
E
G
3Pal
Pip



 98
Suc
E
G
Y(Me)
Pip



 99
Suc
E
G
Pip
Pip



100
Suc
E
G
Tic
Pip



101
Suc
E
G
1Nal
Pip



102
Suc
E
G
F(2F)
Pip



103
Suc
E
G
F(3F)
Pip



104
Suc
E
G
F(4F)
Pip



105
Suc
E
G
F
Tic



106
Suc
E
G
Y
Tic



107
Suc
E
G
W
Tic



108
Suc
E
G
F(3Br)
Tic



109
Suc
E
G
F(4NO2)
Tic



110
Suc
E
G
3Pal
Tic



111
Suc
E
G
Y(Me)
Tic



112
Suc
E
G
Pip
Tic



113
Suc
E
G
Tic
Tic



114
Suc
E
G
1Nal
Tic



115
Suc
E
G
F(2F)
Tic



116
Suc
E
G
F(3F)
Tic



117
Suc
E
G
F(4F)
Tic



118
Suc
E
G
F
1Nal



119
Suc
E
G
Y
1Nal



120
Suc
E
G
W
1Nal



121
Suc
E
G
F(3Br)
1Nal



122
Suc
E
G
F(4NO2)
1Nal



123
Suc
E
G
3Pal
1Nal



124
Suc
E
G
Y(Me)
1Nal



125
Suc
E
G
Pip
1Nal



126
Suc
E
G
Tic
1Nal



127
Suc
E
G
1Nal
1Nal



128
Suc
E
G
F(2F)
1Nal



129
Suc
E
G
F(3F)
1Nal



130
Suc
E
G
F(4F)
1Nal



131
Suc
E
G
F
F(2F)



132
Suc
E
G
Y
F(2F)



133
Suc
E
G
W
F(2F)



134
Suc
E
G
F(3Br)
F(2F)



135
Suc
E
G
F(4NO2)
F(2F)



136
Suc
E
G
3Pal
F(2F)



137
Suc
E
G
Y(Me)
F(2F)



138
Suc
E
G
Pip
F(2F)



139
Suc
E
G
Tic
F(2F)



140
Suc
E
G
1Nal
F(2F)



141
Suc
E
G
F(2F)
F(2F)



142
Suc
E
G
F(3F)
F(2F)



143
Suc
E
G
F(4F)
F(2F)



144
Suc
E
G
F
F(3F)



145
Suc
E
G
Y
F(3F)



146
Suc
E
G
W
F(3F)



147
Suc
E
G
F(3Br)
F(3F)



148
Suc
E
G
F(4NO2)
F(3F)



149
Suc
E
G
3Pal
F(3F)



150
Suc
E
G
Y(Me)
F(3F)



151
Suc
E
G
Pip
F(3F)



152
Suc
E
G
Tic
F(3F)



153
Suc
E
G
1Nal
F(3F)



154
Suc
E
G
F(2F)
F(3F)



155
Suc
E
G
F(3F)
F(3F)



156
Suc
E
G
F(4F)
F(3F)



157
Suc
E
G
F
F(4F)



158
Suc
E
G
Y
F(4F)



159
Suc
E
G
W
F(4F)



160
Suc
E
G
F(3Br)
F(4F)



161
Suc
E
G
F(4NO2)
F(4F)



162
Suc
E
G
3Pal
F(4F)



163
Suc
E
G
Y(Me)
F(4F)



164
Suc
E
G
Pip
F(4F)



165
Suc
E
G
Tic
F(4F)



166
Suc
E
G
1Nal
F(4F)



167
Suc
E
G
F(2F)
F(4F)



168
Suc
E
G
F(3F)
F(4F)



169
Suc
E
G
F(4F)
F(4F)



170
Suc
E(Me)
G
F
F
 (96)


171
Suc
E(Me)
G
Y
F



172
Suc
E(Me)
G
W
F



173
Suc
E(Me)
G
F(3Br)
F



174
Suc
E(Me)
G
F(4NO2)
F



175
Suc
E(Me)
G
3Pal
F



176
Suc
E(Me)
G
Y(Me)
F



177
Suc
E(Me)
G
Pip
F



178
Suc
E(Me)
G
Tic
F



179
Suc
E(Me)
G
1Nal
F



180
Suc
E(Me)
G
F(2F)
F



181
Suc
E(Me)
G
F(3F)
F



182
Suc
E(Me)
G
F(4F)
F



183
Suc
E(Me)
G
F
Y



184
Suc
E(Me)
G
Y
Y



185
Suc
E(Me)
G
W
Y



186
Suc
E(Me)
G
F(3Br)
Y



187
Suc
E(Me)
G
F(4NO2)
Y



188
Suc
E(Me)
G
3Pal
Y



189
Suc
E(Me)
G
Y(Me)
Y



190
Suc
E(Me)
G
Pip
Y



191
Suc
E(Me)
G
Tic
Y



192
Suc
E(Me)
G
1Nal
Y



193
Suc
E(Me)
G
F(2F)
Y



194
Suc
E(Me)
G
F(3F)
Y



195
Suc
E(Me)
G
F(4F)
Y



196
Suc
E(Me)
G
F
W



197
Suc
E(Me)
G
Y
W



198
Suc
E(Me)
G
W
W



199
Suc
E(Me)
G
F(3Br)
W



200
Suc
E(Me)
G
F(4NO2)
W



201
Suc
E(Me)
G
3Pal
W



202
Suc
E(Me)
G
Y(Me)
W



203
Suc
E(Me)
G
Pip
W



204
Suc
E(Me)
G
Tic
W



205
Suc
E(Me)
G
1Nal
W



206
Suc
E(Me)
G
F(2F)
W



207
Suc
E(Me)
G
F(3F)
W



208
Suc
E(Me)
G
F(4F)
W



209
Suc
E(Me)
G
F
F(3Br)



210
Suc
E(Me)
G
Y
F(3Br)



211
Suc
E(Me)
G
W
F(3Br)



212
Suc
E(Me)
G
F(3Br)
F(3Br)



213
Suc
E(Me)
G
F(4NO2)
F(3Br)



214
Suc
E(Me)
G
3Pal
F(3Br)



215
Suc
E(Me)
G
Y(Me)
F(3Br)



216
Suc
E(Me)
G
Pip
F(3Br)



217
Suc
E(Me)
G
Tic
F(3Br)



218
Suc
E(Me)
G
1Nal
F(3Br)



219
Suc
E(Me)
G
F(2F)
F(3Br)



220
Suc
E(Me)
G
F(3F)
F(3Br)



221
Suc
E(Me)
G
F(4F)
F(3Br)



222
Suc
E(Me)
G
F
F(4NO2)



223
Suc
E(Me)
G
Y
F(4NO2)



224
Suc
E(Me)
G
W
F(4NO2)



225
Suc
E(Me)
G
F(3Br)
F(4NO2)



226
Suc
E(Me)
G
F(4NO2)
F(4NO2)



227
Suc
E(Me)
G
3Pal
F(4NO2)



228
Suc
E(Me)
G
Y(Me)
F(4NO2)



229
Suc
E(Me)
G
Pip
F(4NO2)



230
Suc
E(Me)
G
Tic
F(4NO2)



231
Suc
E(Me)
G
1Nal
F(4NO2)



232
Suc
E(Me)
G
F(2F)
F(4NO2)



233
Suc
E(Me)
G
F(3F)
F(4NO2)



234
Suc
E(Me)
G
F(4F)
F(4NO2)



235
Suc
E(Me)
G
F
3Pal



236
Suc
E(Me)
G
Y
3Pal



237
Suc
E(Me)
G
W
3Pal



238
Suc
E(Me)
G
F(3Br)
3Pal



239
Suc
E(Me)
G
F(4NO2)
3Pal



240
Suc
E(Me)
G
3Pal
3Pal



241
Suc
E(Me)
G
Y(Me)
3Pal



242
Suc
E(Me)
G
Pip
3Pal



243
Suc
E(Me)
G
Tic
3Pal



244
Suc
E(Me)
G
1Nal
3Pal



245
Suc
E(Me)
G
F(2F)
3Pal



246
Suc
E(Me)
G
F(3F)
3Pal



247
Suc
E(Me)
G
F(4F)
3Pal



248
Suc
E(Me)
G
F
Y(Me)



249
Suc
E(Me)
G
Y
Y(Me)



250
Suc
E(Me)
G
W
Y(Me)



251
Suc
E(Me)
G
F(3Br)
Y(Me)



252
Suc
E(Me)
G
F(4NO2)
Y(Me)



253
Suc
E(Me)
G
3Pal
Y(Me)



254
Suc
E(Me)
G
Y(Me)
Y(Me)



255
Suc
E(Me)
G
Pip
Y(Me)



256
Suc
E(Me)
G
Tic
Y(Me)



257
Suc
E(Me)
G
1Nal
Y(Me)



258
Suc
E(Me)
G
F(2F)
Y(Me)



259
Suc
E(Me)
G
F(3F)
Y(Me)



260
Suc
E(Me)
G
F(4F)
Y(Me)



261
Suc
E(Me)
G
F
Pip



262
Suc
E(Me)
G
Y
Pip



263
Suc
E(Me)
G
W
Pip



264
Suc
E(Me)
G
F(3Br)
Pip



265
Suc
E(Me)
G
F(4NO2)
Pip



266
Suc
E(Me)
G
3Pal
Pip



267
Suc
E(Me)
G
Y(Me)
Pip



268
Suc
E(Me)
G
Pip
Pip



269
Suc
E(Me)
G
Tic
Pip



270
Suc
E(Me)
G
1Nal
Pip



271
Suc
E(Me)
G
F(2F)
Pip



272
Suc
E(Me)
G
F(3F)
Pip



273
Suc
E(Me)
G
F(4F)
Pip



274
Suc
E(Me)
G
F
Tic



275
Suc
E(Me)
G
Y
Tic



276
Suc
E(Me)
G
W
Tic



277
Suc
E(Me)
G
F(3Br)
Tic



278
Suc
E(Me)
G
F(4NO2)
Tic



279
Suc
E(Me)
G
3Pal
Tic



280
Suc
E(Me)
G
Y(Me)
Tic



281
Suc
E(Me)
G
Pip
Tic



282
Suc
E(Me)
G
Tic
Tic



283
Suc
E(Me)
G
1Nal
Tic



284
Suc
E(Me)
G
F(2F)
Tic



285
Suc
E(Me)
G
F(3F)
Tic



286
Suc
E(Me)
G
F(4F)
Tic



287
Suc
E(Me)
G
F
1Nal



288
Suc
E(Me)
G
Y
1Nal



289
Suc
E(Me)
G
W
1Nal



290
Suc
E(Me)
G
F(3Br)
1Nal



291
Suc
E(Me)
G
F(4NO2)
1Nal



292
Suc
E(Me)
G
3Pal
1Nal



293
Suc
E(Me)
G
Y(Me)
1Nal



294
Suc
E(Me)
G
Pip
1Nal



295
Suc
E(Me)
G
Tic
1Nal



296
Suc
E(Me)
G
1Nal
1Nal



297
Suc
E(Me)
G
F(2F)
1Nal



298
Suc
E(Me)
G
F(3F)
1Nal



299
Suc
E(Me)
G
F(4F)
1Nal



300
Suc
E(Me)
G
F
F(2F)



301
Suc
E(Me)
G
Y
F(2F)



302
Suc
E(Me)
G
W
F(2F)



303
Suc
E(Me)
G
F(3Br)
F(2F)



304
Suc
E(Me)
G
F(4NO2)
F(2F)



305
Suc
E(Me)
G
3Pal
F(2F)



306
Suc
E(Me)
G
Y(Me)
F(2F)



307
Suc
E(Me)
G
Pip
F(2F)



308
Suc
E(Me)
G
Tic
F(2F)



309
Suc
E(Me)
G
1Nal
F(2F)



310
Suc
E(Me)
G
F(2F)
F(2F)



311
Suc
E(Me)
G
F(3F)
F(2F)



312
Suc
E(Me)
G
F(4F)
F(2F)



313
Suc
E(Me)
G
F
F(3F)



314
Suc
E(Me)
G
Y
F(3F)



315
Suc
E(Me)
G
W
F(3F)



316
Suc
E(Me)
G
F(3Br)
F(3F)



317
Suc
E(Me)
G
F(4NO2)
F(3F)



318
Suc
E(Me)
G
3Pal
F(3F)



319
Suc
E(Me)
G
Y(Me)
F(3F)



320
Suc
E(Me)
G
Pip
F(3F)



321
Suc
E(Me)
G
Tic
F(3F)



322
Suc
E(Me)
G
1Nal
F(3F)



323
Suc
E(Me)
G
F(2F)
F(3F)



324
Suc
E(Me)
G
F(3F)
F(3F)



325
Suc
E(Me)
G
F(4F)
F(3F)



326
Suc
E(Me)
G
F
F(4F)



327
Suc
E(Me)
G
Y
F(4F)



328
Suc
E(Me)
G
W
F(4F)



329
Suc
E(Me)
G
F(3Br)
F(4F)



330
Suc
E(Me)
G
F(4NO2)
F(4F)



331
Suc
E(Me)
G
3Pal
F(4F)



332
Suc
E(Me)
G
Y(Me)
F(4F)



333
Suc
E(Me)
G
Pip
F(4F)



334
Suc
E(Me)
G
Tic
F(4F)



335
Suc
E(Me)
G
1Nal
F(4F)



336
Suc
E(Me)
G
F(2F)
F(4F)



337
Suc
E(Me)
G
F(3F)
F(4F)



338
Suc
E(Me)
G
F(4F)
F(4F)



339
Suc
E
G
F
F
 (97)


340
Suc
Q
G
F
F



341
Suc
N
G
F
F



342
Suc
(MeE
G
F
F



343
Suc
βE
G
F
F



344
Suc
(MeD
G
F
F



345
Suc
βD
G
F
F



346
Suc
E(Me
G
F
F



347
Suc
D
G
F
F



348
Suc
Gla
G
F
F



349
Suc
E
G
Y
F



350
Suc
Q
G
Y
F



351
Suc
N
G
Y
F



352
Suc
(Me)E
G
Y
F



353
Suc
βE
G
Y
F



354
Suc
(Me)D
G
Y
F



355
Suc
βD
G
Y
F



356
Suc
E(Me)
G
Y
F



357
Suc
D
G
Y
F



358
Suc
Gla
G
Y
F



359
Suc
E
G
W
F



360
Suc
Q
G
W
F



361
Suc
N
G
W
F



362
Suc
(Me)E
G
W
F



363
Suc
βE
G
W
F



364
Suc
(Me)D
G
W
F



365
Suc
βD
G
W
F



366
Suc
E(Me)
G
W
F



367
Suc
D
G
W
F



368
Suc
Gla
G
W
F



369
Suc
E
G
F(3Br)
F



370
Suc
Q
G
F(3Br)
F



371
Suc
N
G
F(3Br)
F



372
Suc
(Me)E
G
F(3Br)
F



373
Suc
βE
G
F(3Br)
F



374
Suc
(Me)D
G
F(3Br)
F



375
Suc
βD
G
F(3Br)
F



376
Suc
E(Me)
G
F(3Br)
F



377
Suc
D
G
F(3Br)
F



378
Suc
Gla
G
F(3Br)
F



379
Suc
E
G
F(4NO2)
F



380
Suc
Q
G
F(4NO2)
F



381
Suc
N
G
F(4NO2)
F



382
Suc
(Me)E
G
F(4NO2)
F



383
Suc
βE
G
F(4NO2)
F



384
Suc
(Me)D
G
F(4NO2)
F



385
Suc
βD
G
F(4NO2)
F



386
Suc
E(Me)
G
F(4NO2)
F



387
Suc
D
G
F(4NO2)
F



388
Suc
Gla
G
F(4NO2)
F



389
Suc
E
G
3Pal
F



390
Suc
Q
G
3Pal
F



391
Suc
N
G
3Pal
F



392
Suc
(Me)E
G
3Pal
F



393
Suc
βE
G
3Pal
F



394
Suc
(Me)D
G
3Pal
F



395
Suc
βD
G
3Pal
F



396
Suc
E(Me)
G
3Pal
F



397
Suc
D
G
3Pal
F



398
Suc
Gla
G
3Pal
F



399
Suc
E
G
Y(Me)
F



400
Suc
Q
G
Y(Me)
F



401
Suc
N
G
Y(Me)
F



402
Suc
(Me)E
G
Y(Me)
F



403
Suc
βE
G
Y(Me)
F



404
Suc
(Me)D
G
Y(Me)
F



405
Suc
βD
G
Y(Me)
F



406
Suc
E(Me)
G
Y(Me)
F



407
Suc
D
G
Y(Me)
F



408
Suc
Gla
G
Y(Me)
F



409
Suc
E
G
Pip
F



410
Suc
Q
G
Pip
F



411
Suc
N
G
Pip
F



412
Suc
(Me)E
G
Pip
F



413
Suc
βE
G
Pip
F



414
Suc
(Me)D
G
Pip
F



415
Suc
βD
G
Pip
F



416
Suc
E(Me)
G
Pip
F



417
Suc
D
G
Pip
F



418
Suc
Gla
G
Pip
F



419
Suc
E
G
Tic
F



420
Suc
Q
G
Tic
F



421
Suc
N
G
Tic
F



422
Suc
(Me)E
G
Tic
F



423
Suc
βE
G
Tic
F



424
Suc
(Me)D
G
Tic
F



425
Suc
βD
G
Tic
F



426
Suc
E(Me)
G
Tic
F



427
Suc
D
G
Tic
F



428
Suc
Gla
G
Tic
F



429
Suc
E
G
1Nal
F



430
Suc
Q
G
1Nal
F



431
Suc
N
G
1Nal
F



432
Suc
(Me)E
G
1Nal
F



433
Suc
βE
G
1Nal
F



434
Suc
(Me)D
G
1Nal
F



435
Suc
βD
G
1Nal
F



436
Suc
E(Me)
G
1Nal
F



437
Suc
D
G
1Nal
F



438
Suc
Gla
G
1Nal
F



439
Suc
E
G
F(2F)
F



440
Suc
Q
G
F(2F)
F



441
Suc
N
G
F(2F)
F



442
Suc
(Me)E
G
F(2F)
F



443
Suc
βE
G
F(2F)
F



444
Suc
(Me)D
G
F(2F)
F



445
Suc
βD
G
F(2F)
F



446
Suc
E(Me)
G
F(2F)
F



447
Suc
D
G
F(2F)
F



448
Suc
Gla
G
F(2F)
F



449
Suc
E
G
F(3F)
F



450
Suc
Q
G
F(3F)
F



451
Suc
N
G
F(3F)
F



452
Suc
(Me)E
G
F(3F)
F



453
Suc
βE
G
F(3F)
F



454
Suc
(Me)D
G
F(3F)
F



455
Suc
βD
G
F(3F)
F



456
Suc
E(Me)
G
F(3F)
F



457
Suc
D
G
F(3F)
F



458
Suc
Gla
G
F(3F)
F



459
Suc
E
G
F(4F)
F



460
Suc
Q
G
F(4F)
F



461
Suc
N
G
F(4F)
F



462
Suc
(Me)E
G
F(4F)
F



463
Suc
βE
G
F(4F)
F



464
Suc
(Me)D
G
F(4F)
F



465
Suc
βD
G
F(4F)
F



466
Suc
E(Me)
G
F(4F)
F



467
Suc
D
G
F(4F)
F



468
Suc
Gla
G
F(4F)
F



469
Suc
E
G
F
F
 (98)


470
Suc
Q
G
F
F



471
Suc
N
G
F
F



472
Suc
(Me)E
G
F
F



473
Suc
βE
G
F
F



474
Suc
(Me)D
G
F
F



475
Suc
βD
G
F
F



476
Suc
E(Me)
G
F
F



477
Suc
D
G
F
F



478
Suc
Gla
G
F
F



479
Suc
E
G
F
Y



480
Suc
Q
G
F
Y



481
Suc
N
G
F
Y



482
Suc
(Me)E
G
F
Y



483
Suc
βE
G
F
Y



484
Suc
(Me)D
G
F
Y



485
Suc
βD
G
F
Y



486
Suc
E(Me)
G
F
Y



487
Suc
D
G
F
Y



488
Suc
Gla
G
F
Y



489
Suc
E
G
F
W



490
Suc
Q
G
F
W



491
Suc
N
G
F
W



492
Suc
(Me)E
G
F
W



493
Suc
βE
G
F
W



494
Suc
(Me)D
G
F
W



495
Suc
βD
G
F
W



496
Suc
E(Me)
G
F
W



497
Suc
D
G
F
W



498
Suc
Gla
G
F
W



499
Suc
E
G
F
F(3Br)



500
Suc
Q
G
F
F(3Br)



501
Suc
N
G
F
F(3Br)



502
Suc
(Me)E
G
F
F(3Br)



503
Suc
βE
G
F
F(3Br)



504
Suc
(Me)D
G
F
F(3Br)



505
Suc
βD
G
F
F(3Br)



506
Suc
E(Me)
G
F
F(3Br)



507
Suc
D
G
F
F(3Br)



508
Suc
Gla
G
F
F(3Br)



509
Suc
E
G
F
F(4NO2)



510
Suc
Q
G
F
F(4NO2)



511
Suc
N
G
F
F(4NO2)



512
Suc
(Me)E
G
F
F(4NO2)



513
Suc
βE
G
F
F(4NO2)



514
Suc
(Me)D
G
F
F(4NO2)



515
Suc
βD
G
F
F(4NO2)



516
Suc
E(Me)
G
F
F(4NO2)



517
Suc
D
G
F
F(4NO2)



518
Suc
Gla
G
F
F(4NO2)



519
Suc
E
G
F
3Pal



520
Suc
Q
G
F
3Pal



521
Suc
N
G
F
3Pal



522
Suc
(Me)E
G
F
3Pal



523
Suc
βE
G
F
3Pal



524
Suc
(Me)D
G
F
3Pal



525
Suc
βD
G
F
3Pal



526
Suc
E(Me)
G
F
3Pal



527
Suc
D
G
F
3Pal



528
Suc
Gla
G
F
3Pal



529
Suc
E
G
F
Y(Me)



530
Suc
Q
G
F
Y(Me)



531
Suc
N
G
F
Y(Me)



532
Suc
(Me)E
G
F
Y(Me)



533
Suc
βE
G
F
Y(Me)



534
Suc
(Me)D
G
F
Y(Me)



535
Suc
βD
G
F
Y(Me)



536
Suc
E(Me)
G
F
Y(Me)



537
Suc
D
G
F
Y(Me)



538
Suc
Gla
G
F
Y(Me)



539
Suc
E
G
F
Pip



540
Suc
Q
G
F
Pip



541
Suc
N
G
F
Pip



542
Suc
(Me)E
G
F
Pip



543
Suc
βE
G
F
Pip



544
Suc
(Me)D
G
F
Pip



545
Suc
βD
G
F
Pip



546
Suc
E(Me)
G
F
Pip



547
Suc
D
G
F
Pip



548
Suc
Gla
G
F
Pip



549
Suc
E
G
F
Tic



550
Suc
Q
G
F
Tic



551
Suc
N
G
F
Tic



552
Suc
(Me)E
G
F
Tic



553
Suc
βE
G
F
Tic



554
Suc
(Me)D
G
F
Tic



555
Suc
βD
G
F
Tic



556
Suc
E(Me)
G
F
Tic



557
Suc
D
G
F
Tic



558
Suc
Gla
G
F
Tic



559
Suc
E
G
F
1Nal



560
Suc
Q
G
F
1Nal



561
Suc
N
G
F
1Nal



562
Suc
(Me)E
G
F
1Nal



563
Suc
βE
G
F
1Nal



564
Suc
(Me)D
G
F
1Nal



565
Suc
βD
G
F
1Nal



566
Suc
E(Me)
G
F
1Nal



567
Suc
D
G
F
1Nal



568
Suc
Gla
G
F
1Nal



569
Suc
E
G
F
F(2F)



570
Suc
Q
G
F
F(2F)



571
Suc
N
G
F
F(2F)



572
Suc
(Me)E
G
F
F(2F)



573
Suc
βE
G
F
F(2F)



574
Suc
(Me)D
G
F
F(2F)



575
Suc
βD
G
F
F(2F)



576
Suc
E(Me)
G
F
F(2F)



577
Suc
D
G
F
F(2F)



578
Suc
Gla
G
F
F(2F)



579
Suc
E
G
F
F(3F)



580
Suc
Q
G
F
F(3F)



581
Suc
N
G
F
F(3F)



582
Suc
(Me)E
G
F
F(3F)



583
Suc
βE
G
F
F(3F)



584
Suc
(Me)D
G
F
F(3F)



585
Suc
βD
G
F
F(3F)



586
Suc
E(Me)
G
F
F(3F)



587
Suc
D
G
F
F(3F)



588
Suc
Gla
G
F
F(3F)



589
Suc
E
G
F
F(4F)



590
Suc
Q
G
F
F(4F)



591
Suc
N
G
F
F(4F)



592
Suc
(Me)E
G
F
F(4F)



593
Suc
βE
G
F
F(4F)



594
Suc
(Me)D
G
F
F(4F)



595
Suc
βD
G
F
F(4F)



596
Suc
E(Me)
G
F
F(4F)



597
Suc
D
G
F
F(4F)



598
Suc
Gla
G
F
F(4F)



599
Suc
E
G
F
F
 (99)


600
Peg35
E
G
F
F



601
Peg30
E
G
F
F



602
Taa
E
G
F
F



603
Caa
E
G
F
F



604
Fum
E
G
F
F



605
TA
E
G
F
F



606
Ia
E
G
F
F



607
1,4-Chda
E
G
F
F



608
1,2-Chda
E
G
F
F



609
Ga
E
G
F
F



610
Suc
E
G
Y
F



611
Peg35
E
G
Y
F



612
Peg30
E
G
Y
F



613
Taa
E
G
Y
F



614
Caa
E
G
Y
F



615
Fum
E
G
Y
F



616
TA
E
G
Y
F



617
Ia
E
G
Y
F



618
1,4-Chda
E
G
Y
F



619
1,2-Chda
E
G
Y
F



620
Ga
E
G
Y
F



621
Suc
E
A
F
F
(100)


622
Peg35
E
A
F
F



623
Peg30
E
A
F
F



624
Taa
E
A
F
F



625
Caa
E
A
F
F



626
Fum
E
A
F
F



627
TA
E
A
F
F



628
Ia
E
A
F
F



629
1,4-Chda
E
A
F
F



630
1,2-Chda
E
A
F
F



631
Ga
E
A
F
F



632
Suc
E
a
F
F



633
Peg35
E
a
F
F



634
Peg30
E
a
F
F



635
Taa
E
a
F
F



636
Caa
E
a
F
F



637
Fum
E
a
F
F



638
TA
E
a
F
F



639
Ia
E
a
F
F



640
1,4-Chda
E
a
F
F



641
1,2-Chda
E
a
F
F



642
Ga
E
a
F
F



643
Suc
E
r3A
F
F



644
Peg35
E
r3A
F
F



645
Peg30
E
r3A
F
F



646
Taa
E
r3A
F
F



647
Caa
E
r3A
F
F



648
Fum
E
r3A
F
F



649
TA
E
r3A
F
F



650
Ia
E
r3A
F
F



651
1,4-Chda
E
r3A
F
F



652
1,2-Chda
E
βA
F
F



653
Ga
E
βA
F
F



654
Suc
E
P
F
F



655
Peg35
E
P
F
F



656
Peg30
E
P
F
F



657
Taa
E
P
F
F



658
Caa
E
P
F
F



659
Fum
E
P
F
F



660
TA
E
P
F
F



661
Ia
E
P
F
F



662
1,4-Chda
E
P
F
F



663
1,2-Chda
E
P
F
F



664
Ga
E
P
F
F



665
Suc
E
P
F
F



666
Peg35
E
p
F
F



667
Peg30
E
p
F
F



668
Taa
E
p
F
F



669
Caa
E
p
F
F



670
Fum
E
p
F
F



671
TA
E
p
F
F



672
Ia
E
p
F
F



673
1,4-Chda
E
p
F
F



674
1,2-Chda
E
p
F
F



675
Ga
E
p
F
F



676
Suc
E
Sar
F
F



677
Peg35
E
Sar
F
F



678
Peg30
E
Sar
F
F



679
Taa
E
Sar
F
F



680
Caa
E
Sar
F
F



681
Fum
E
Sar
F
F



682
TA
E
Sar
F
F



683
Ia
E
Sar
F
F



684
1,4-Chda
E
Sar
F
F



685
1,2-Chda
E
Sar
F
F



686
Ga
E
Sar
F
F



687
Suc
E
β-H-ala
F
F



688
Peg35
E
β-H-ala
F
F



689
Peg30
E
β-H-ala
F
F



690
Taa
E
β-H-ala
F
F



691
Caa
E
β-H-ala
F
F



692
Fum
E
β-H-ala
F
F



693
TA
E
β-H-ala
F
F



694
Ia
E
β-H-ala
F
F



695
1,4-Chda
E
β-H-ala
F
F



696
1,2-Chda
E
β-H-ala
F
F



697
Ga
E
β-H-ala
F
F



698
Suc
E
Ahx
F
F



699
Peg35
E
Ahx
F
F



700
Peg30
E
Ahx
F
F



701
Taa
E
Ahx
F
F



702
Caa
E
Ahx
F
F



703
Fum
E
Ahx
F
F



704
TA
E
Ahx
F
F



705
Ia
E
Ahx
F
F



706
1,4-Chda
E
Ahx
F
F



707
1,2-Chda
E
Ahx
F
F



708
Ga
E
Ahx
F
F



709
Suc
E
F
F




710
Peg35
E
F
F




711
Peg30
E
F
F




712
Taa
E
F
F




713
Caa
E
F
F




714
Fum
E
F
F




715
TA
E
F
F




716
Ia
E
F
F




717
1,4-Chda
E
F
F




718
1,2-Chda
E
F
F




719
Ga
E
F
F




720
Suc
E
F
F
E



721
Suc
E
Ahx
E




722
Suc
E
Ahx
F
E
(101)


723
Suc
E
Ahx
F
Y



724
Suc
E
Ahx
F
W



725
Suc
E
Ahx
F
F(3Br)



726
Suc
E
Ahx
F
F(4NO2)



727
Suc
E
Ahx
F
3Pal



728
Suc
E
Ahx
F
Y(Me)



729
Suc
E
Ahx
F
Pip



730
Suc
E
Ahx
F
Tic



731
Suc
E
Ahx
F
1Nal



732
Suc
E
Ahx
F
F(2F)



733
Suc
E
Ahx
F
F(3F)



734
Suc
E
Ahx
F
F(4F)



735
Suc
E
Ahx
F
F
(102)


736
Suc
Q
Ahx
F
F



737
Suc
N
Ahx
F
F



738
Suc
(Me)E
Ahx
F
F



739
Suc
βE
Ahx
F
F



740
Suc
(Me)D
Ahx
F
F



741
Suc
βD
Ahx
F
F



742
Suc
E(Me)
Ahx
F
F



743
Suc
D
Ahx
F
F



744
Suc
Gla
Ahx
F
F



745
Ga
E
G
F
Y(Me)
(103)


746
Suc
E
Ahx
Y
F
(104)


747
Suc
E
Ahx
W
F



748
Suc
E
Ahx
F(3Br)
F



749
Suc
E
Ahx
F(4NO2)
F



750
Suc
E
Ahx
3Pal
F



751
Suc
E
Ahx
Y(Me)
F



752
Suc
E
Ahx
Pip
F



753
Suc
E
Ahx
Tic
F



754
Suc
E
Ahx
1Nal
F



755
Suc
E
Ahx
F(2F)
F



756
Suc
E
Ahx
F(3F)
F



757
Suc
E
Ahx
F(4F)
F



758
Suc
E
Ahx
F
F



759
Suc
Ahx
G
F
F
(105)


760
Suc
E
Ahx
F
F
(106)


761
Suc
E
G
Ahx
F
(107)


762
Suc
E
G
F
Ahx
(108)


763
Suc
E
F
F
E
(109)


764
Peg35
E
F
F
E



765
Peg30
E
F
F
E



766
Taa
E
F
F
E



767
Caa
E
F
F
E



768
Fum
E
F
F
E



769
TA
E
F
F
E



770
Ia
E
F
F
E



771
1,4-Chda
E
F
F
E



772
1,2-Chda
E
F
F
E



773
Ga
E
F
F
E



774
Suc
E
Y
F
E



775
Peg35
E
Y
F
E



776
Peg30
E
Y
F
E



777
Taa
E
Y
F
E



778
Caa
E
Y
F
E



779
Fum
E
Y
F
E



780
TA
E
Y
F
E



781
Ia
E
Y
F
E



782
1,4-Chda
E
Y
F
E



783
1,2-Chda
E
Y
F
E



784
Ga
E
Y
F
E



785
Peg35
E
F
Y(Me)
E



786
Peg30
E
F
Y(Me)
E



787
Taa
E
F
Y(Me)
E



788
Caa
E
F
Y(Me)
E



789
Fum
E
F
Y(Me)
E



790
TA
E
F
Y(Me)
E



791
Ia
E
F
Y(Me)
E



792
1,4-Chda
E
F
Y(Me)
E



793
1,2-Chda
E
F
Y(Me)
E



794
Ga
E
F
Y(Me)
E





Ahx = aminohexanoic acid






Selected peptides from the arrays shown in Tables 5 and 6 were re-synthesised and analysed using the FP assay described above. The results of this assay are shown in Table 7.









TABLE 7







FP assay results of selected peptides











FP Assay


Entry
Sequence
IC50/μM





 1
1,4-Chda-EGFFE-NH2
>100   





 2
EAFFE-NH2
  28   





 3
Ga-EGFFE-NH2
  65   





 4
Suc-EG-1Nal-F(4NO2)-E-NH2
  68   





 5
Suc-EG-1Nal-Y(Me)-E-NH2
   5.4 





 6
Suc-EG-F(2F)-F(3F)-NH2
>100   





 7
Suc-EG-F(2F)-F(4NO2)-E-NH2
   4.9 





 8
Suc-EG-F(2F)-Y(4Me)-E-NH2
   3.2 





 9
Suc-EG-F(3F)-3Pal-NH2
  70   





10
Suc-EG-F(3F)-F(3F)-NH2
  44   





11
Suc-EG-F(3F)-F(4NO2)-E-NH2
   0.52





12
Suc-EG-F(3F)-F(4NO2)-NH2
>100   





13
Suc-EG-F(4Br)-F(3F)-NH2
  81   





14
Suc-EG-F(4Br)-F-NH2
  38   





15
Suc-EG-F(4Br)-F-NH2
  39   





16
Suc-EG-F(4F)-F(4NO2)-E-NH2
   1.2 





17
Suc-EG-F(4NO2)-1Nal-NH2
  14   





18
Suc-EG-F(4NO2)-F(4NO2)-NH2
  78   





19
Suc-EGF-F(3F)-NH2
>100   





20
Suc-EGF-F(4F)-NH2
>100   





21
Suc-EGF-F(4NO2)-NH2
>100   





22
Suc-EGFF-NH2
  46   





23
Suc-EGF-Y(Me)-E(OMe)-NH2
  71   





24
Suc-EGY-F(3F)-NH2
  91   





25
Suc-EGY-F(4F)-NH2
  71   





26
Suc-EGY-F(4NO2)-NH2
  91   





27
Suc-EGYFE-NH2
   1   





28
Suc-EGYF-NH2
  31   





29
Suc-QGYF-NH2
  49   





30
Suc-βE-GYFE-NH2
  30   









X-EGXXE-NH2 was identified as a useful consensus binding motif and further peptides were designed and synthesised to increase potency. These peptides were tested in the FP assay described above and the results are shown in Table 8.









TABLE 8







Further modification of


peptide sequence X-EGXXE-NH2











FP Assay


Entry
Sequence (SEQ ID NO:)
IC50/μM












 1
(MeO)Suc-EG-F(3F)-1Nal-E-NH2 (110)
1.6





 2
2-NaphthylSO2-dEG-F(3F)-WE-NH2
0.044





 3
3,4-(MeO)2PhSO2-dEG-F(3F)-WE-NH2
0.042





 4
4-(BuO)PhSO2-dEG-F(3F)-WE-NH2
0.056





 5
4-(MeO)PhSO2-dEG-F(3F)-WE-NH2
0.017





 6
4-(MeO)PhSO2-DEG-F(3F)-WE-NH2 (111)
0.027





 7
4-(PhO)PhSO2-dEG-F(3F)-WE-NH2
0.063





 8
Ac-dEG-F(3F)-1Nal-E-NH2
0.112





 9
Ac-dEG-F(3F)-WE-NH2
0.102





10
AG-F(3F)-F(4NO2)-E-NH2 (112)
>100





11
AGYFE-NH2 (112)
>100





12
Bz-dEG-F(3F)-WE-NH2
0.173





13
EA-F(3F)-F(4NO2)-E-NH2 (113)
>100





14
EAFFE-NH2 (113)
>100





15
EAYFE-NH2 (113)
>100





16
EG-F(3F)-1Nal-E-NH2 (113)
>100





17
EG-F(4NO2)-1Nal-E-NH2 (113)
>100





18
EGY-1Nal-E-NH2 (113)
>100





19
EGY-F(4NO2)-E-NH2 (113)
>100





20
EtCO-dEG-F(3F)-WE-NH2
0.145





21
EtOCO-dEG-F(3F)-WE-NH2
0.045





22
Fum-EG-F(3F)-F(4NO2)-E-NH2 (114)
60.4





23
Mal-EG-F(3F)-F(4NO2)-E-NH2 (114)
2.17





24
MeoCO-dEG-F(3F)-WE-NH2
0.076





25
QG-F(3F)-F(4NO2)-E-NH2 (115)
>100





26
QGFFE-NH2 (115)
>100





27
QGYFE-NH2 (115)
>100





28
Suc-AG-F(3F)-F(4NO2)-E-NH2 (116)
8.01





29
Suc-AGYFE-NH2 (116)
22.2





30
Suc-EA-F(3F)-F(4NO2)-E-NH2 (117)
27.5





31
Suc-EAFFE-NH2 (117)
59.8





32
Suc-EAYFE-NH2 (117)
82.1





33
Suc-EG-3Pal-1Nal-E-NH2 (117)
3.63





34
Suc-EG-F(3F)-1Nal-E-NH2 (117)
0.157





35
Suc-EG-F(3F)-1Nal-Q-NH2 (117)
7.22





36
Suc-EG-F(3F)-HE-NH2 (117)
0.521





37
Suc-EG-F(3F)-WE-NH2 (117)
0.095





38
Suc-EG-F(3F)-Y(Me)-E-NH2 (117)
1.4





39
Suc-EG-F(4NO2)-1Nal-E-NH2 (117)
1.31





40
Suc-EGI-1Nal-E-NH2 (117)
15.09





41
Suc-EGY-1Na1-E-NH2 (117)
1.15





42
Suc-EGY-F(4NO2)-E-NH2 (117)
2.07





43
Suc-QG-F(3F)-F(4NO2)-E-NH2 (115)
2.56





44
Suc-QGFFE-NH2 (115)
10





45
Suc-QGYFE-NH2 (115)
10.7





46
Ts-dDG-F(3F)-WD-NH2
0.081





47
Ts-dDG-F(3F)-WE-NH2
0.025





48
Ts-dEGF(3Cl)WE(Me)-NH2
0.017





49
Ts-dEGF(3Cl)WE-NH2
0.01





50
Ts-dEG-F(3F)-WD-NH2
0.034





51
Ts-dEGF(3F)W-E(Me)-NH2
0.022





52
Ts-dEG-F(3F)-WE-NH2
0.029





53
Ts-DEG-F(3F)-WE-NH2
0.018









Example 2—in Silico Biotin Capture Assay of Lead Test Compounds

A non-fluorescent based assay was used to validate potential binding peptides. The phosphopeptide substrate KKERLLDDRHDpSGLDpSMKDEE was biotinylated by coupling a biotinamido-hexanoic acid succinimide ester to a lysine in the peptide. 0.5 μg of each protein was mixed with 50 picomoles of biotinylated peptide in a total volume of 50 μl. The buffer conditions were 50 mM Hepes 7.5 and 100 mM NaCl. Compounds were then added to a final concentration of 60 μM. The reaction was allowed to incubate for 1 hour at room temperature and then 5 μl of streptavidin-agarose beads was added and incubated for 1 hour. Beads were washed twice with buffer and run on an SDS-PAGE gel. Proteins were visualized by anti-His antibody. The results of this assay are shown in FIG. 6.


Example 3—Compound Hits from In-Vitro Assays Tested with SPR

Binding of SucEGF(4NO2)1NalENH2 to βTrCP1 was tested using the Surface Plasmon Resonance (SPR) method described above.


Example 4—Measurement of Inhibition of the E3 Ligase Cascades βTrCP(IκBα) and βTrCP(β-Catenin)

Candidate compounds were tested in duplicate at both 10 and 100 μM against the E3 ligase cascades βTrCP(IκBα) and βTrCP(β-catenin). The E3 assays were carried out according to the component concentrations detailed in Table 9. In each case the E2 was HA,6His-UbcH3-(hu,FL), the E1 was 6His-UBE1-(hu,FL) and the ubiquitin (Sigma U6253) was biotinylated at a 5:1 ratio. The E3 tetramer constructs and substrate pairings are shown in Table 10. Substrate phosphorylation was performed in the absence of compound; consequently any observed signal modulation should not reflect inhibition of the up-stream kinase reaction. All other steps (E1, E2 and substrate ubiquitination) were carried out in the presence of compound. The stopped reaction mix (10 μl) was added to an ECL plate loaded with anti c-Myc ( 1/500 Dilution, Millipore 05-724) and blocked with 5% BSA. Binding was allowed to proceed for 1 hour at RT before a wash step (3 40 μl PBST washes). Detection was achieved by binding SA-Ru TAG at 1 μg/ml (1 hour @ RT, 3 40 μl PBST washes) before reading on an MSD Sector Imager 6000.









TABLE 9







Assay concentrations










βTrCP(IκBα)
βTrCP(β-catenin)

















Ub
2
μM
2
μM



ATP
10
μM
10
μM



E2
250
nM
250
nM



E1
2.5
nM
2.5
nM



E3 Tetramer
0.3
μg/well
0.3
μg/well



Sub
200
nM
200
nM

















TABLE 10







E3 Ligase and substrate pairings








E3 Tetramer
Substrate





GST-βTrCP1-(iso1,hu,53-end,E353D)/GST-Skp1-
cMyc, 6His-IκBα-


(hu,fl)/6His-Cul1-(hu,fl)/UT-Rbx1-(hu,fl)
(hu,FL)


GST-βTrCP1-(iso1,hu,53-end,E353D)/GST-Skp1-
cMyc,6His-β-


(hu,fl)/6His-Cul1-(hu,fl)/UT-Rbx1-(hu,fl)
catenin-(hu,FL)









Assay results are shown in FIG. 8. Assays were performed as described above


Example 5—Selectivity Analysis of Top Compounds—FP Against Fbw 7

Candidate compounds were tested for inhibition of Fbw7 and Skp2 as described above. As shown in FIGS. 9A-9B, none of them showed any activity above background at concentrations of 10 μM or 100 μM.


Example 6—Collation of Data Collected on Candidate Compounds

Table 11 (below) shows the collation of all data points collected for the most promising compounds.









TABLE 11







Summary of peptide data

















Biotin








Pulldown

% Inhibition


















Assay
Ub Assay
Skp2 @
Fbw7 @




FP
SPR
10 μM/
10 μM/
10 μM
10 μM


Entry
Sequence
IC50/μM
Kd/μM
100 μM
100 μM
(100 μM)
(100 μM)





 1
DpSGIFE-NH2
 1.24 ± 0.145
 1.477 ± 0.286
reduces/
reduces/
 8 (13)
 0 (0)






inhibits
inhibits







 2
Suc-EGFFE-NH2
 3.18 ± 0.357
29.456 ± 4.613
no effect/
reduces/
14 (0)
 0 (0)






reduces
inhibits







 3
Suc-EGF(2F)F(4NO2)E-NH2
 4.91 ± 0.702
 8.000 ± 0.260
no effect/
no effect/
 8 (11)
 0 (0)






reduces
inhibits







 4
Suc-EGF(3F)F(4NO2)E-NH2
0.522 ± 0.049
 4.530 ± 0.681
reduces/
inhibits/
 0 (3)
 0 (2)






inhibits
inhibits







 5
Suc-EGF(4F)F(4NO2)E-NH2
 1.21 ± 0.093
 0.507 ± 0.267
no effect/
reduces/
 0 (0)
 0 (0)






inhibits
inhibits







 6
Suc-EGF(2F)Y(Me)E-NH2
 3.28 ± 0.500
 4.400 ± 0.820
no effect/
no effect/
 0 (4)
 0 (4)






reduces
inhibits







 7
Suc-EGYFE-NH2
 1.04 ± 0.101
 0.537 ± 0.117
no effect/
reduces/
11 (28)
 0 (0)






inhibits
inhibits







 8
Mal-EGF(3F)F(4NO2)E-NH2
 2.17 ± 0.425
 1.463 ± 0.035
no effect/
no effect/
 0 (14)
 0 (18)






inhibits
inhibits







 9
Suc-EGY1NalE-NH2
 1.15 ± 0.222
 1.490 ± 0.625
no effect/
reduces/
 0 (6)
 1 (5)






inhibits
inhibits







10
SucEGF(3F)1NalE-NH2
0.157 ± 0.037
 0.320 ± 0.240
inhibits/
inhibits/
 0 (12)
 3 (0)






inhibits
inhibits







11
Suc-EGF(4NO2)1NalE-NH2
 1.31 ± 0.198
 1.965 ± 0.635
reduces/
reduces/
12 (6)
14 (28)






inhibits
inhibits







12
Suc-QGF(3F)F(4NO2)E-NH2
 2.56 ± 0.374
 5.491 ± 2.271
no effect/
no effect/
 0 (0)
 0 (0)






reduces
inhibits







13
Suc-EGYF(4NO2)E-NH2
 2.07 ± 0.346
17.713 ± 5.196
no effect/
no effect/
11 (31)
 0 (0)






inhibits
inhibits







14
Suc-EGF(3F)WE-NH2
0.095 ± 0.009
 0.455 ± 0.030
reduces/
reduces/
 5 (6)
 0 (0)






inhibits
inhibits







15
Suc-EGF(3F)HE-NH2
0.521 ± 0.059
 0.400 ± 0.021
reduces/
reduces/
 3 (18)
12 (83)






inhibits
inhibits







16
Ac-dEGF(3F)1NalE-NH2
0.112 ± 0.017
 0.590 ± 0.282
reduces/
reduces/
 0 (7)
 3 (8)






inhibits
inhibits







17
Ac-dEGF(3F)WE-NH2
0.102 ± 0.017
 0.250 ± 0.059
reduces/
reduces/
10 (27)
 3 (0)






inhibits
inhibits







18
Bz-dEGF(3F)WE-NH2
0.173 ± 0.072
0.4240 ± 0.067
reduces/
reduces/
 0 (0)
 0 (5)






inhibits
inhibits







19
Et(CO)-dEGF(3F)WE-NH2
0.145 ± 0.026
 0.150 ± 0.065
reduces/
reduces/
 1 (10)
 0 (10)






inhibits
inhibits







20
MeO(CO)-dEGF(3F)WE-NH2
0.076 ± 0.014
 0.053 ± 0.015
reduces/
inhibits/
 0 (16)
 6 (8)






inhibits
inhibits







21
Ts-dEGF(3F)WE-NH2
0.029 ± 0.005
 0.190 ± 0.042
reduces/
inhibits/
19 (25)
 9 (0)






inhibits
inhibits







22
4-(MeO)-PhSO2-
0.017 ± 0.002
 0.048 ± 0.003
s. reduces/
inhibits/
 7 (12)
 0 (18)



dEGF(3F)WE-NH2


inhibits
inhibits







23
EtO(CO)-dEGF(3F)WE-NH2
0.045 ± 0.005
 0.034 ± 0.017
reduces/
inhibits/
 4 (4)
 0 (12)






inhibits
inhibits







24
Ts-DEGF(3F)WE-NH2
0.018 ± 0.002
 0.019 ± 0.013
s. reduces/
inhibits/
 0 (7)
 0 (8)






inhibits
inhibits







25
Ts-dDGF(3F)WE-NH2
0.025 ± 0.004
 0.292 ± 0.106
s. reduces/
inhibits/
 0 (22)
 1 (8)






inhibits
inhibits







26
4-(MeO)-PhSO2-
0.027 ± 0.002
 0.495 ± 0.163
s. reduces/
inhibits/
15 (3)
 0 (8)



DEGF(3F)WE-NH2


inhibits
inhibits







27
Ts-dEGF(3F)WD-NH2
0.034 ± 0.005
 0.680 ± 0.152
s. reduces/
inhibits/
 4 (6)
 3 (8)






inhibits
inhibits







28
Ts-dDGF(3F)WD-NH2
0.081 ± 0.020
 0.637 ± 0.114
reduces/
inhibits/
 0 (14)
 4 (10)






inhibits
inhibits







29
3,4-(MeO)2-PhSO2-
0.042 ± 0.001
 0.603 ± 0.144
reduces/
reduces/
19 (19)
 2 (0)



dEGF(3F)WE-NH2


inhibits
inhibits







30
4-(BuO)-PhSO2-
0.056 ± 0.008
 0.767 ± 0.009
reduces/
reduces/
 0 (13)
 7 (12)



dEGF(3F)WE-NH2


inhibits
inhibits







31
2-NaphthylSO2-
0.044 ± 0.005
 0.262 ± 0.021
reduces/
reduces/
 0 (17)
10 (11)



dEGF(3F)WE-NH2


inhibits
inhibits







32
Ts-dEGF(3F)WE(Me)-NH2
0.022 ± 0.003
 0.170 ± 0.092
s. reduces/
inhibits/
 0 (0)
 4 (10)






inhibits
inhibits







33
Ts-dEGF(3Cl)WE(Me)-NH2
0.017 ± 0.003
 0.047 ± 0.025
s. reduces/
inhibits/
 0 (10)
 7 (11)






inhibits
inhibits







34
Ts-dEGF(3Cl)WE-NH2
 0.01 ± 0.002
 0.154 ± 0.057
s. reduces/
inhibits/
 0 (0)
 8 (6)






inhibits
inhibits









Example 7—Cell-Based Activity of Peptidomimetics

The activity of 4-(MeO)-PhSO2-dEGF(3F)WE-NH2 in a cell was investigated as an example of activity seen with this family of compounds.


In-Cell Western Assay for PDCD4 Accumulation.


PDCD4 is a substrate of βTrCP and so an inhibitor of βTrCP should result in the stabilisation and accumulation of PDCD4 in cells. To measure this an in-cell western assay was used and the peptidomimetics were delivered to the cells by nucleofection.


Nucleofection


MCF7 cells were grown in 10 cm dishes in 10 ml DMEM+10% FBS and 1% Pen/Strep at 37° C./5% CO2. On the day of nucleofection, the dish was washed with 90% confluent MCF7 cells with 5 ml PBS, then 3 ml of Trypsin/EDTA was added and incubated at 37° C./5% CO2 for approximately 5 mins until the cells detached from the plastic. 7 ml of normal growth media was added and the number of cells present was counted using a haemocytometer.


The cells were centrifuged cells at 90 g for 10 mins at RT and the supernatant was removed. 100 μl of Nucleofection Buffer V+ Supplement was added (Lonza Biologics Cat no VCA-1003) per 8×105 cells, and the cells were added to peptidomimetics dissolved in <3% DMSO.


The cell/peptidomimetic mix was added to cuvettes supplied for the nucleofector and the sample was nucleofected using the recommended MCF7 programme for high cell viability (i.e. E-014). 500 μl of TPA (12-O-tetradecanoylphorbol-13-acetate)-supplemented growth media (i.e. DMEM/10% FBS/1% Pen-Strep/10 nM TPA) was added to the cuvette and 100 μl of this solution was transferred to a well of a 96 well plate and incubated at 37° C./5% CO2 for 8 hours.


In Cell Western


The celled were fixed by removing the growth media, adding 3.7% Formaldehyde in PBS and incubating at RT for 20 mins. The plate was washed three times in PBS. And the cells were permeabilised by washing 5× for 5 mins each in PBS+0.1% Triton X-100.


The cells were blocked by adding 3% BSA in PBS-Tween to the cells and incubating at RT for 1.5 hours. An anti-PDCD4 antibody (Abcam cat no: ab80590) was added at a concentration of 1:1000 in 3% BSA in PBS-T (50 ul/well) and incubated at RT for 2.5 hours. The cells were washed 5× for 5 mins each with PBS-T before the secondary antibody (LiCor Biosciences Donkey Anti-Rabbit IRDye 800CW cat no 926-32213) was added at a concentration of 1:1000, along with the DNA stain DRAQ5 at a concentration of 1:10000, in 3% BSA in PBS-T (50 ul/well) and incubated at RT for 1 hour, protected from light.


The cells were washed cells 5× for 5 mins each with PBS-T and all the liquid was removed from the wells before the plate was read on the LiCor Biosciences Odyssey at 700 nm and 800 nm. The reading was normalised in the 800 nm channel to that in the 700 nm channel. The data are shown in FIG. 12, where DMSO was used as a negative control and 20 μM MG132 was used as a positive control. The data is presented as percentage activity with DMSO=0% and 20 M MG132=100% activity. Error bars represent standard deviation from 3 replicates.


Fluorescence Reporter Assay for PDCD4 Accumulation


This assay uses two stable cell lines expressing PDCD4 fused to a GFP tag. One cell line (MCF7:GFP-PDCD4WT) shows an increase in nuclear fluorescence when βTrCP is inhibited due to the accumulation of GFP-PDCD4 in the nucleus. The other cell line (MCF7:GFP-PDCD4S71A/S76A or or MCF7:GFP-PDCD4Mut) does not show an increase in nuclear fluorescence when βTrCP is inhibited because of a mutation of two serine residues in the phosphodegron of PDCD4 that are required for βTrCP recognition. This allows false positives to be identified, where accumulation of PDCD4 is not due to stabilisation by inhibiting βTrCP.


Nucleofection


The two stable cell lines MCF7:GFP-PDCD4WT and MCF7:GFP-PDCD4S71A/S76Awere grown in 10 ml DMEM+10% FBS and 1% Pen/Strep supplemented with 2 mg/ml Geneticin at 37° C./5% CO2. These cell lines were nucleofected as described above except for the additional supplementation of all media with 2 mg/ml Geneticin.


Fluorescent Reporter Assay


The cells were fixed by removing the growth media and adding 3.7% Formaldehyde in PBS to the cells. The cells were then incubated at RT for 20 mins and the plates washed 3× in PBS.


DRAQ5 DNA stain was added to the cells in PBS at a concentration of 1:10000 and incubated at RT for 1 hour protecting from light. The plate was washed 3× in PBS and read on the Perkin Elmer OPERA platform using the nuclei counting algorithm F in both the 488 nm channel (GFP) and 640 nm channel (DRAQ5). The number of GFP-positive nuclei was expressed as a percentage of total number of nuclei, and the data are shown in FIG. 13. DMSO was used as a negative control and 10 μM MG132 was used as a positive control. Error bars represent standard deviation from 6 replicates. The data are expressed as percentage activity with DMSO (WT)=0% and 10 μM MG132 (WT)=100% activity.


Example 8—Cell Based Activity of Cell Permeable Compounds

The compounds shown in FIGS. 14A-14B were synthesised to improve cell permeability. FIG. 15 shows the accumulation of PDCD4 as measured by the in cell western assay described above, following treatment of MCF7 cells with UBP036. FIG. 16 compares the round II compounds (shown in FIGS. 14A-14B) with UBP036 in this assay. FIG. 17 shows the accumulation of β-catenin, a further substrate of βTrCP, as measured by the in cell western assay following treatment of MCF7 cells with UBP036. Inhibition of βTrCP, which is responsible for the degradation of both PDCD4 and β-catenin, causes a concomitant stabilisation and accumulation of levels of these proteins which can be measured by in cell western assay.



FIG. 18 shows the accumulation of PDCD4 as measured by the fluorescence reporter assay described in example 7. There is accumulation of GFP-PDCD4WT, while GFP-PDCD4Mut, which had been mutated such that the interaction between PDCD4 and βTrCP is abolished, shows no accumulation or stabilisation. This confirms that the stabilisation resulting from the compounds is dependent on the βTrCP/PDCD4 interaction and is not due to another mechanism such as a global increase in protein production.


Testing of UBP037 and UBP038 by in cell western provided inconclusive results. The fluorescence readings were consistently under those of the DMSO negative control. This suggested that the compounds were interfering in some way with the fluorescence readout assay. When these compounds were tested by traditional western blot however, they exhibited cellular activity. The results for PDCD4 accumulation in MCF7 cells are shown in FIG. 19. The results for PDCD4 accumulation in LNCaP cells are shown in FIG. 20. The western blot and in cell western are based on the same scientific principles and differ only in the technology used. It was concluded that UBP037 and UBP038 are incompatible with the in cell western technology, but the western blot data shows that these compounds inhibit βTrCP.


β-Catenin in Cell Western


Plating of the MCF7 cells onto 96 well plates—this step is carried out 1 day before the treatment of the cells to allow the cells to adhere well to the tissue culture plastic. MCF7 cells to be used for seeding should be less than 100% confluent in a 10 cm dish. Add 3 ml of RT trypsin/EDTA to the cells. Incubate at 37° C./5% CO2 for a few minutes until the cells easily come away from the plastic by gentle swirling. Add 7 ml of media to the cells. Wash the bottom of the plate gently with the 10 ml to ensure all cells are captured and dispense into a 15 ml falcon. Add 1001l of the cells to 1001l of Trypan blue and add to haemocytometer to count the number of cells present. Make up approx 10 ml of cells in media at 2×104 cells/1001l (well) with fresh media. Add this correct seeding density to a 10 cm dish. Dispense 1001l of cells/well into a 96 well plate without using the outside wells. Incubate at 37° C./5% CO2 overnight


Treatment of MCF7 cells with compound of interest (COI) and controls—Put OptiMEM into 37° C. water bath to warm. Add 4 ul of 50 mM compound X for testing to eppendorf—this is tube 1. Add 2 al DMSO to 6 tubes marked tube 2-7. Take 21 al of 50 mM (COI) and add to 2 al of DMSO in tube 2 and pipette up and down to mix. Take 21 al from tube 2 and add to 2 μl DMSO in next tube and mix. Repeat until 2 μl in all 7 tubes and have 1:2 serial dilutions from tube 1 to 7 (final conc.: 250 PM to 2 μM). Add 3.5 μl DMSO in tube marked “DMSO” (final percentage 0.5% as for all compounds). Add 0.7 μl of 10 mM MG132 and 2.8pal DMSO in tube marked “MG132” (final conc.: 10 μM). Add 0.7pal of 10 mM MLN4924 and 2.8 μM DMSO in tube marked “MLN4924” (final conc. 10 uM). Add 4001l of pre-warmed OptiMEM to each of tubes 1-7 with serial dilution of compound X. Add 700 ul of pre-warmed OptiMEM to tubes marked “DMSO”, “MG132” and “MLN4924”. Take plate with seeded MCF7 cells and remove the media from the first column of wells. Add 100 μl of “DMSO” to each of six wells in first column of wells. Remove media from the second column of wells and add 100 μl of “MG132” to each of six wells in second column of wells. Remove media from the third column of wells and add 100 μl of “MLN4924” to each of six wells in third column of wells. Remove media from the remaining columns of wells in small batches and add 100 μl of each dilution of compound X to three wells in fourth to tenth column of wells. Incubate at 37° C./5% CO2 for 8 hrs. Add 1 ml of 37% formaldehyde to 9 ml PBS. Remove the media from the plate and add 100 μl of 3.7% formaldehyde to each well. Incubate at RT for 20 mins. Remove formaldehyde and add 100 μl of PBS to each well. Remove PBS and add another 100 μl PBS to each well. Store at 4° C. overnight.


Immunostaining of β-catenin using In Cell Western (ICW) protocol—Add 100 μl of PBS+0.1% triton to each well and incubate at RT with gentle mixing for 5 mins. Replace with fresh PBS+0.1% triton and repeat 4 times. Note—all washing steps must be carried out gently to avoid dislodging cells. Add 100 μl of 3% BSA in PBS-Tween to each well and incubate at RT with gentle mixing for 1 hour. Add 6.51 al of β-catenin antibody to 6.5 ml of 3% BSA in PBS-Tween. Add 100 μl of primary antibody solution to all bar one of the DMSO-treated wells. This will act as a negative control. It is also possible to not add primary antibody to one well of each treated column of wells in order to have a no primary control for all positive controls and each concentration of COI. Incubate at RT with gentle mixing for 2.5 hours or overnight at 4° C. Add 100 μl of PBS-Tween to each well and incubate at RT with gentle mixing for 5 mins. Replace with fresh PBS-Tween and repeat 4 times. Spin down the vial of anti-rabbit IR800 (LI-COR Biosciences: cat no 926-32213 Donkey Anti-Rabbit IRDye 800CW) at top speed for a few seconds and add 6.5 μl of this to 6.5 ml of 3% BSA in PBS-Tween. Add 50 μl of this secondary antibody solution to one well as a control for DRAQ5. Add 0.65pal of DRAQ5 to the secondary antibody solution and add 100 μl of this to all other wells. Incubate at RT with gentle mixing for 1 hr with the plates protected from light with tin foil. Add 100 μl of PBS-Tween to each well and incubate at RT with gentle mixing for 5 mins. Replace with fresh PBS-Tween and repeat 4 times


Traditional Western Blot

    • Seed MCF7 or LNCaP cells in 12 well plates at 100,000cells/well in complete medium (RPMI 1640, 10% fetal bovine serum, 100 units/mL penicillin, 100 ug/mL streptomycin, and 2 mmol/L glutamine, all from GIBCO) and incubate at 37° C./5% CO2 overnight.
    • 24 hrs later change media to complete media+compound of interest+10 nM TPA and incubate for 8 hrs
    • After incubation, wash cells three times in cold PBS and lyse with 3T3 lysis buffer containing protease inhibitors (Roche).
    • Protein quantification was determined by the Bradford assay (BIORAD).
    • Run 5 ug of protein samples on 4-12% NuPAGE Bis-Tris gels (Invitrogen) and transfer onto nitrocellulose membrane (Whatman).
    • Block membranes in 50% PBS, 50% Odyssey block (LI-COR) for 1 hour.
    • Incubate blots with primary antibodies diluted in 49% PBST, 49% Odyssey block and 0.5% 10% Tween-20 (Sigma) overnight at the following concentrations:
      • Anti-PDCD4 (Abcam) at 1/10000
      • Anti-Tubulin (Abcam) at 1/20000
      • Anti-Actin (Abcam) at 1/5000.
    • Wash blots 3 times for 5 min in PBST.
    • Incubate with secondary antibodies on blots for 1 hour, protected from light.
      • Antibodies were diluted in the same solution as primary antibodies
      • 1/20000 goat anti-mouse alexa 680 (LI-COR),
      • 1/10000 donkey anti-rabbit alexa 800 (LI-COR).
    • Wash blots 3 times for 5 min in PBST.
    • Observe protein bands using LI-COR Odyssey reader and quantitate strength of bands with Odyssey software.


Example 9—Cell Based Activity of Cell Permeable Compounds on the xCELLigence Platform

The effects of the compounds shown in FIGS. 14A-14B were tested on cancer cell lines MCF7 (breast cancer) and LNCaP (prostate cancer) using the xCELLigence platform (http://www.roche-applied-science.com/sis/xcelligence/ezhome.html). This method of measuring cell viability via measuring cell index is known in the art.


Different doses of the compounds were tested on MCF7 cells to see if there would be an effect on their cell viability. FIG. 21 shows that increasing compound concentration is concomitant with decreased cell viability. These experiments were repeated with LNCaP cells as shown in FIG. 22 (B).


In order to prove that this activity was specific to the active compounds of the invention, a control compound was developed that is identical to the active compound except for the amino acid sequence that confers the specificity of the active compound. The control compound was compared to its partner active compound (UBP037). FIG. 22 (C) shows that the loss in cell viability (LNCaP cells) seen with the active species is not seen with the control compound.


xCELLigence Method

    • Before starting the experiment add 50 μL of medium (RPMI 1640, 10% fetal bovine serum, 100 units/mL penicillin, 100 ug/mL streptomycin, and 2 mmol/L glutamine, all from GIBCO) to each well of 96 well E-plate (Roche) and place the plate in the xCELLigence platform to measure the background.
    • LNCaP/PNT-1 cells are seeded (LNCaPs 80000 cells/well; PNT1 3000 cells/well) in the plate in complete medium and equilibrated for 60 mins at RT before returning to the xCELLigence platform and incubating at 37° C./5% CO2.
    • 24 hrs after seeding, remove 100 μL of medium was removed from each well and cells were treated with compounds or DMSO diluted in 100 μL of OptiMEM (GIBCO).
    • Replace the plate in the xCELLigence platform
    • Cells growth can then be monitored in real-time by the cell index profile on the xCELLigence readout screen
    • Cell growth can be expressed as normalized cell index and doubling time/slope calculated using the RTCA software.


Example 10—Activity in Cancer Cells Lines Compared to Non-Cancer Cell Lines

The susceptibility of cancer cell lines versus non-cancer cell lines following treatment with the compounds was also investigated. FIG. 23 shows that UBP036 and UBP037 and UPB038 inhibit the growth of the cancer cell line LNCaP, while having far less effect on the non-malignant PNT-1 cell line.


Analysis


1. Demonstration of Cell Based Activity by Active Compounds in Comparison to an Inactive Compound of a Similar Physicochemical Nature


This has been demonstrated by the xCELLigence assay that compared UBP037 with the control compound (Ts-EdFEGW-Ahx-K(Stearic)-NH2, a scrambled version of a compound of the present invention (see FIG. 22 (C)). UBP037 severely restricts the proliferation of the LNCaP cells, while the control compound shows levels of cell proliferation similar to that seen with DMSO. Therefore, the activity seen in UBP037 is entirely due to the active peptide moiety and not the delivery vehicle (stearic acid). In addition, UBP036, UBP022, UBP090 all display cell based activity. The delivery vehicle in all these examples is very different (cholesterol, poly-lysine, Log P manipulation) with the only common feature being the active species. If cell based activity is due to an off-target effect, it is highly unlikely that all three vehicles would hit the same non-specific target. Finally the potency of these cell active species is exactly that seen with the “naked” active peptide upon nucleofection into the cells (see FIG. 24) and thus any off-target effect on the same range of biomarkers seen by three separate CPP delivery moieties would be highly unlikely.


2. Demonstration of Target-Specific Activity in a Cell Based Assay Format


This is illustrated for compound UBP036 when examined in the GFP reporter (FIG. 18). When PDCD4 is mutated to eliminate the PDCD4-βTrCP interaction, there is no accumulation of PDCD4 in response to treatment with UBP036. This implies that any increase in PDCD4 seen in the wild type construct is entirely dependent on the interaction between βTrCP and PDCD4.


In addition, the use of two βTrCP substrates; PDCD4 and 1-catenin, in the in cell western assay would also suggest that any effect seen is due to βTrCP inhibition.


Again the active species of all the compounds in FIG. 14 is identical, the only difference between them being the cell delivery vehicle. As discussed, there appears to be no activity associated with the cell-delivery vehicle, and therefore the PDCD4 biomarker activity and cellular proliferation activity seen for all the compounds is βTrCP-dependent.


3. Demonstration of Activity in Several Cancer Cell Lines


In the development of the assays for a βTrCP inhibitor a number of cell-line —biomarker combinations were surveyed to identify the most sensitive assay for βTrCP inhibitors. The breast cancer cell-line MCF7 proved extremely responsive to βTrCP inhibition and this could be measured by the rapid and robust accumulation of the βTrCP substrates PDCD4 and 1-catenin.


As can been seen from the data presented here all compounds exhibit this activity (to various degrees) in MCF7 cells.


The next phase of development involved addressing the potential therapeutic benefit of βTrCP inhibition in a cell line that could be replicated in an animal model. Here LNCaPs were chosen given previous evidence that βTrCP inhibition inhibited cell proliferation both in vitro and in vivo (PLoS One. 2010 Feb. 5; 5(2):e9060.) Again it can be seen from the data that all compounds tested exhibit a reduction in proliferation (to various degrees) in LNCaP cells.


In addition to the cellular activity seen in each of these cell lines, it is also becoming apparent that the biomarker activity assays in MCF7 cells appear to predict the therapeutic activity seen in LNCaP cells.


There is also cell viability inhibition in MCF7 cells upon treatment with these compounds (see FIGS. 22A-E) revealing the possibility of further therapeutic uses for 3 TrCP inhibitors in a breast cancer model (and a potential mechanism of action in PDCD4 accumulation).


Medical Applications of βTrCP Inhibitors


There are a number of potential indications for βTrCP inhibitors including many forms of cancer.


Two key indications exemplified are prostate cancer and breast cancer.


Breast Cancer.


There is clinical evidence that βTrCP2 is over expressed in a number of cancers including breast cancer [J Biol Chem. 2002, 277, 36624-30]. This study also demonstrated that the cell lines used to model breast cancer such as MCF7 cells also display this same overexpression when compared to non-cancer cell lines such as MCF10A. This implies that work done on βTrCP inhibition in these cancer cell lines could indeed reflect potential outcomes in a clinical setting.


There have been numerous in vivo studies to demonstrate the importance of βTrCP in mammary development. In βTrCP1−/− mice there is a hypoplastic phenotype observed where cell proliferation is reduced by 50% in the mammary gland with other organs unaffected. Furthermore, when there is exogenous high expression of βTrCP1 introduced in the mammary epithelia, approx 40% of mice develop carcinomas. [Mol Cell Biol. 2004, 24, 8184-94.]. The value of this study is two-fold. It demonstrates that despite the widespread expression of βTrCP, a systemic reduction in βTrCP levels (via the genetic ablation of βTrCP1) has a preferential effect on the mammary gland. Also it reveals that overexpression of βTrCP1 can in itself result in an increased cancer risk in this tissue. This suggests inhibition of βTrCP may be of value in both breast cancers that do not display βTrCP overexpression (as inhibition of βTrCP in healthy animals appears to preferentially target the mammary gland for reduced cell proliferation) and those that do (due to the potential causative effect of βTrCP mis-regulation).


In addition to the value of inhibiting βTrCP alone to affect favourable outcomes in breast cancer, there is also work to suggest that combining βTrCP inhibition with some of the current therapies for breast cancer could improve the outcome of these therapies. Inhibition of βTrCP by an RNAi approach suppressed growth and survival of human breast cancer cells [Cancer Res. 2005 Mar. 1; 65(5):1904-8]. It is worth noting that these experiments were carried out on both ER-positive and ER-negative breast cancer cell lines with βTrCP inhibition having a similar impact on both. In addition, inhibition of βTrCP augmented the anti-proliferative effects of anticancer drugs such as doxorubicin, tamoxifen, and paclitaxel on human breast cancer cells. These data suggest that βTrCP inhibition could be effective as a front line adjuvant therapy or in combination with an existing breast cancer treatment regime.


We have shown that the βTrCP inhibitors described here inhibit binding of βTrCP to IκBα (a well-known βTrCP substrate) in in vitro binding assays and stabilise several βTrCP substrates in cell based assays. In addition this inhibitor can reduce the cell viability of a breast cancer cell line in a similar fashion to βTrCP RNAi.


These data and the studies described above show that βTrCP is a validated, novel target in breast cancer, and that its inhibition is tractable and of clinical significance.


Prostate Cancer.


Here the main evidence for the role of βTrCP is from the work of Yinon Ben-Neriah and Eli Pikarsky [PLoS One. 2010 Feb. 5; 5(2):e9060.] Their key finding here was not only that inhibition of βTrCP resulted in the loss of cell viability of LNCaP cells in vitro—but also that when this inhibition is transferred from cells to animals through the use of LNCaP xenografts—it results in a loss of growth of prostate tumours and in combination with androgen ablation—the lack of tumour growth entirely.


Example 11—Effect of Capping Groups on Peptide Activity

A number of different C-terminal and N-terminal capping groups were added to peptide d-E-G-F(3F)—W-E-NH2 in order to demonstrate how the inhibitory activity of d-E-G-F(3F)—W-E-NH2 is affected by particular capping groups, which act to increase cell penetration (as illustrated by Δc Log P, relative to UBP022). Ki and Δc Log P values for the the capped compounds are shown in Table 12.









TABLE 12







Ki and ΔcLogP for N- and C-terminal


capped peptide d-E-G-F(3F)-W-E-NH2













Average




Code
Sequence
Ki/nM
ΔcLogP
Kd/ΔcLogP





UBP022
4-(MeO)PhSO2-dEGF(3F)WE-NH2
  5.775







UBP054
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(t-Bu-Ph))-NH2
  1.938
3.84
  0.505





UBP055
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-2-Naph)-NH2
  1.851
3.51
  0.527





UBP056
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(2,4,6-(Me)3-Ph))-NH2
  3.387
3.27
  1.036





UBP057
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(ME)-Ph))-NH2
  2.884
2.64
  1.092





UBP058
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Br)-Ph))-NH2
  3.3455
3.03
  1.104





UBP059
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-(Br)-Ph))-NH2
  2.960
2.40
  1.2333





UBP060
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(Cl)-Ph))-NH2
  3.641
2.94
  1.238





UBP061
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCOPh)-NH2
  3.105
2.33
  1.333





UBP062
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(3,5-(Cl)2-Ph))-NH2
  6.229
3.55
  1.755





UBP063
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(CH2)4CH3)-NH2
  5.374
2.71
  1.983





UBP064
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(CF3)-Ph))-NH2
  6.391
3.09
  2.068





UBP065
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-Ph)-NH2
  5.939
2.55
  2.329





UBP066
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-(4-(OMe)-Ph))-NH2
  5.1745
2.22
  2.331





UBP068
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCONH-Ph)-NH2
  5.608
2.22
  2.526





UBP068
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHCO-CH2CH2Ch(CH3)2)-NH2
  5.378
2.12
  2.537





UBP069
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHOCO-1-Naph)-NH2
 11.608
3.73
  3.112





UBP070
4-(MeO)PhSO2 dEGF(3F)WE-Ahx-K(NHCO-(4-(Cl)-2,6(Fl2-Ph))-NH2
 11.455
3.06
  3.743





UBP071
4-(MeO)PhSO2 dEGF(3F)WE-Ahx-K(NHCO-(4-(Me2N)-Ph))-NH2
  9.506
2.33
  4.080





UBP072
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-i-Pr)-Ph))-NH2
 12.941
2.80
  4.622





UBP073
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2Ph)-NH2
 13.005
1.70
  7.65





UBP074
4-(MeO)PhSO2-dEGF(3F)WE-Ahx-K(NHSO2-(4-(n-Pr)-Ph)-NH2
 28.585
2.84
 10.055





UBP040
4-i-Pr-PhSO2-dEGF(3F)WE-NH2
  1.603
1.20
  1.336





UBP041
4-Pr-PhSO2-dEGF(3F)WE-NH2
  1.768
1.24
  1.426





UBP042
4-Br-PhSO2-dEGF(3F)WE-NH2
  2.519
0.8
  3.14





UBP043
4-Br-2-CH3-PhSO2-dEGF(3F)WE-NH2
  3.961
1.12
  3.537





UBP044
2-Naph-SO2-dEGF(3F)WE-NH2
  5.814
1.29
  4.507





UBP045
4-OCF3-PhSO2-dEGF(3F)WE-NH2
  4.774
0.94
  5.079





UBP046
4-Br-3-CF3-PhSO2-dEGF(3F)WE-NH2
 14.217
1.57
  9.055





UBP047
4-CF3-PhSO2-dEGF(3F)WE-NH2
  9.450
0.94
 10.862





UBP048
2,4-(Cl)2-PhSO2-dEGF(3F)WE-NH2
 27.025
1.33
 20.320





UBP049
2,4-(Br)2-PhSO2-dEGF(3F)WE-NH2
 45.255
1.50
 30.17





UBP050
3,5-(CH3)2-PhSO2-dEGF(3F)WE-NH2
 24.645
0.74
 33.30





UBP051
4-Br-2-OCF3-PhSO2-dEGF(3F)WE-NH2
109.55
1.63
 67.209





UBP052
4-I-PhSO2-dEGF(3F)WE-NH2
252.9
1.04
243.17





UBP053
4-Cl-PhSO2-dEGF(3F)WE-NH2
330.5
0.72
459.0









As suggested by the data in table 12, modification of the C-terminal capping group has little effect upon activity. Modification of the N-terminal capping group has a more profound effect. The function of the capping groups is to aid cell penetration as demonstrated by the Δc Log P values. c Log P values are calculated by means well known to the person skilled in the art.


REFERENCES



  • Pons et al. (2008) Biochemistry 47, pg. 14-29

  • Tapia et al. (2008) J. Pept. Sci. 14, pg. 1309-1314

  • Rautio et al. (2008) Nat. Rev. Drug Discov. 7, pg 255-270. using anti-His and anti-βTrCP antibodies



Remington's Pharmaceutical Sciences



  • Stocks et al. (2007) On Medicinal Chemistry

  • Werle et al. (1997) British Journal of Cancer

  • Bungaard et al. Design of Prodrugs

  • Ornstein et al. (1993) Bioorg. Med. Chem. Lett

  • Lakshmann et al (2008) Expert Opinion in Therapeutic Targets 12(7):855-870.

  • Frescas and Pagano (2008) Nature Reviews Cancer Jun; 8(6):438-49

  • Nalepa, Rolfe and Harper (2006). Nature Reviews Drug Discovery 5:596-613

  • Crosetto, Bienko and Dikic (2006) Molecular Cancer Research 4(12): 899-904



SEQUENCES











(consensus sequence)



SEQ ID NO: 1



XXGFXX







(preferred peptide)



SEQ ID NO: 2



dEGF(3F)WE







(preferred peptide)



SEQ ID NO: 3



DEGF(3F)WE







(preferred peptide)



SEQ ID NO: 4



DEGF(3F)WD







(preferred peptide)



SEQ ID NO: 5



dDGF(3F)WD







(preferred peptide)



SEQ ID NO: 6



EGF(3F)WE







(preferred peptide)



SEQ ID NO: 7



dEGF(3F)1NalE







(preferred peptide)



SEQ ID NO: 8



EGF(3F)1NalE







(phosphopeptide substrate)



SEQ ID NO: 9



KKERLLDDRHDpSGLDpSMKDEE







(optimisation starting sequence)



SEQ ID NO: 10



LDpSGIHS







(Vpu phosphodegeneron)



SEQ ID NO: 11



DpSGIHS







(binding peptide)



SEQ ID NO: 12



DpSGIFE







(binding peptide)



SEQ ID NO: 13



DEGIFE







(binding peptide)



SEQ ID NO: 14



ERAEDpSGNEpSEGEIS







(binding peptide)



SEQ ID NO: 15



ERAEDAGNEpSEGEIS







(binding peptide)



SEQ ID NO: 16



ERAEDpSGNEpSEGEHS







(binding peptide)



SEQ ID NO: 17



ERADDpSGNEpSEGEIS







(binding peptide)



SEQ ID NO: 18



dEGIFE







(binding peptide)



SEQ ID NO: 19



dEGIFD







(binding peptide)



SEQ ID NO: 20



dNGIFR







(binding peptide)



SEQ ID NO: 21



DEGFFE







(binding peptide)



SEQ ID NO: 22



dNGFFR







(binding peptide)



SEQ ID NO: 23



EGIFE







(binding peptide)



SEQ ID NO: 24



EGFFE







(binding peptide)



SEQ ID NO: 25



DEGYFE







(binding peptide)



SEQ ID NO: 26



EpSGIFE







(binding peptide)



SEQ ID NO: 27



DpSGIFH







(binding peptide)



SEQ ID NO: 28



DpSGNFE







(binding peptide)



SEQ ID NO: 29



DDpSSGIHS







(binding peptide)



SEQ ID NO: 30



LDpSSGIHS







(binding peptide)



SEQ ID NO: 31



GDpSGIHS







(binding peptide)



SEQ ID NO: 32



ADpSGIHS







(binding peptide)



SEQ ID NO: 33



VDpSGIHS







(control peptide)



SEQ ID NO: 34



DAGIFE







(control peptide)



SEQ ID NO: 35



AGIFE







(control peptide)



SEQ ID NO: 36



AGFFE







(control peptide)



SEQ ID NO: 37



dAGIFR







(control peptide)



SEQ ID NO: 38



dAGIFD







(control peptide)



SEQ ID NO: 39



dAGIFE







(control peptide)



SEQ ID NO: 40



DAGFFE







(control peptide)



SEQ ID NO: 41



DAGYFE







(control peptide)



SEQ ID NO: 42



EAGIFE







(control peptide)



SEQ ID NO: 43



DAGIFH







(control peptide)



SEQ ID NO: 44



DAGNFE







(control peptide)



SEQ ID NO: 45



DAGIHS







(control peptide)



SEQ ID NO: 46



DDASGIHS







(control peptide)



SEQ ID NO: 47



LDASGIHS







(control peptide)



SEQ ID NO: 48



GDAGIHS







(control peptide)



SEQ ID NO: 49



ADAGIHS







(control peptide)



SEQ ID NO: 50



VDAGIHS







(binding peptide)



SEQ ID NO: 51



EGF(3F)HE-NH2







(binding peptide)



SEQ ID NO: 52



dEGF(3F)1Nal-E-NH2







(binding peptide)



SEQ ID NO: 53



EGI1NalE-NH2







(binding peptide)



SEQ ID NO: 54



EGF(3F)1NalQ-NH2







(binding peptide)



SEQ ID NO: 55



EGF(3F)Y(4Me)E-NH2







(binding peptide)



SEQ ID NO: 56



dEGF(3F)WD-NH2





Claims
  • 1.-107. (canceled)
  • 108. A compound of formula IAAA,
  • 109. The compound of claim 108, wherein R4 is independently —CO2H or —CH2CO2H and RA3 is selected from the group consisting of —F, —Cl, —Br, —I.
  • 110. The compound of claim 108, wherein R4 is independently —CO2H or —CH2CO2H; RA3 is selected from the group consisting of —F, —Cl, —Br, —I; and R3 is
  • 111. The compound of claim 108, wherein R4 is independently —CH2CO2H; RA3 is selected from the group consisting of —F, —Cl, —Br, —I; and R3 is
  • 112. The compound of claim 108 wherein R4 is independently —CH2CO2H, RA3 is —F, and R3 is
  • 113. The compound of claim 108, wherein the CG and/or the optional capping group is selected from the group consisting of:
  • 114. The compound of claim 108, wherein the CG and/or the optional capping group is selected from List 1:
  • 115. The compound of claim 108, wherein RA3 is selected from the group consisting of —F, —OH and —NO2.
  • 116. The compound of claim 108, wherein RA3 is —F at one or more of positions 2, 3, 4.
  • 117. The compound of claim 108, wherein RA3 is —Cl at one or more of positions 2, 3, 4.
  • 118. A pharmaceutical composition comprising the compound of claim 108.
  • 119. A method of treating a disease associated with aberrant protein degradation comprising administering the compound of claim 108.
  • 120. A diagnostic kit comprising the compound of claim 108.
Priority Claims (1)
Number Date Country Kind
1110938.6 Jun 2011 GB national
CROSS REFERENCE TO OTHER APPLICATIONS

This application is a continuation of U.S. application Ser. No. 14/127,430, filed Jun. 30, 2014, which is a national stage of PCT Application No. PCT/GB2012/051507, filed on Jun. 27, 2012. This application claims priority under 35 USC § 119 to British Application No. 1110938.6, filed on Jun. 27, 2011.

Continuations (1)
Number Date Country
Parent 14127430 Jun 2014 US
Child 16129700 US