PEPTIDE LIGANDS FOR BINDING TO EphA2

Information

  • Patent Application
  • 20210261620
  • Publication Number
    20210261620
  • Date Filed
    June 18, 2019
    5 years ago
  • Date Published
    August 26, 2021
    2 years ago
Abstract
A peptide ligand specific for EphA2 comprising a polypeptide comprising three residues selected from cysteine, L-2,3-diaminopropionic acid (Dap), N-beta-alkyl-L-2,3-diaminopropionic acid (N-AlkDap) and N-beta-haloalkyl-L-2,3-diaminopropionic acid (N-HAlkDap), with the proviso that at least one of said three residues is selected from Dap, N-AlkDap or N-HAlkDap, the said three residues being separated by at least two loop sequences, and a molecular scaffold, the peptide being linked to the scaffold by covalent alkylamino linkages with the Dap or N-AlkDap or N-HAlkDap residues of the polypeptide and by thioether linkages with the cysteine residues of the polypeptide when the said three residues include cysteine, such that two polypeptide loops are formed on the molecular scaffold.
Description
TECHNICAL FIELD

The present invention relates to peptide ligands showing high binding affinity to the Eph receptor tyrosine kinase A2 (EphA2). The invention also includes drug conjugates comprising said peptides, conjugated to one or more effector and/or functional groups, to pharmaceutical compositions comprising said peptide ligands and drug conjugates and to the use of said peptide ligands and drug conjugates in preventing, suppressing or treating a disease or disorder characterised by overexpression of EphA2 in diseased tissue (such as a tumour).


In particular, the invention relates to peptide ligands of this type having novel chemistries for forming two or more bonds between a peptide and a scaffold molecule.


BACKGROUND OF THE INVENTION

Different research teams have previously tethered peptides to scaffold moieties by forming two or more thioether bonds between cysteine residues of the peptide and suitable functional groups of a scaffold molecule. For example, methods for the generation of candidate drug compounds by linking cysteine-containing peptides to a molecular scaffold as for example tris(bromomethyl) benzene are disclosed in WO 2004/077062 and WO 2006/078161.


The advantage of utilising cysteine thiols for generating covalent thioether linkages in order to achieve cyclisation resides is their selective and biorthogonal reactivity. Thiol-containing linear peptides may be cyclised with a thiol-reactive scaffold compound such as 1, 3, 5 tris-bromomethylbenzene (TBMB) to form Bicyclic Peptides, and the resultant product contains three thioethers at the benzylic locations. The overall reaction of the linear peptide with TBMB to form a looped bicyclic peptide with thioether linkages is shown in FIG. 1.


A need exists for alternative chemistries for coupling peptides to scaffold moieties to form looped peptide structures employing suitable replacements of the thioether moiety, thereby achieving compatibility with different peptides, changes in physiochemical properties such as improved solubility, changes in biodistribution and other advantages.


WO2011/018227 describes a method for altering the conformation of a first peptide ligand or group of peptide ligands, each peptide ligand comprising at least two reactive groups separated by a loop sequence covalently linked to a molecular scaffold which forms covalent bonds with said reactive groups, to produce a second peptide ligand or group of peptide ligands, comprising assembling said second derivative or group of derivatives from the peptide(s) and scaffold of said first derivative or group of derivatives, incorporating one of: (a) altering at least one reactive group; or (b) altering the nature of the molecular scaffold; or (c) altering the bond between at least one reactive group and the molecular scaffold; or any combination of (a), (b) or (c).


Our earlier pending applications PCT/EP2017/083953 and PCT/EP2017/083954 filed 20th Dec. 2017 describe bicycle peptides in which one or more thioether linkages to the scaffold molecule have been replaced by alkylamino linkages.


Eph receptor tyrosine kinases (Ephs) belong to a large group of receptor tyrosine kinases (RTKs), kinases that phosphorylate proteins on tyrosine residues. Ephs and their membrane bound ephrin ligands (ephrins) control cell positioning and tissue organization (Poliakov et al. (2004) Dev Cell 7, 465-80). Functional and biochemical Eph responses occur at higher ligand oligomerization states (Stein et al. (1998) Genes Dev 12, 667-678).


Among other patterning functions, various Ephs and ephrins have been shown to play a role in vascular development. Knockout of EphB4 and ephrin-B2 results in a lack of the ability to remodel capillary beds into blood vessels (Poliakov et al., supra) and embryonic lethality. Persistent expression of some Eph receptors and ephrins has also been observed in newly-formed, adult micro-vessels (Brantley-Sieders et al. (2004) Curr Pharm Des 10, 3431-42; Adams (2003) J Anat 202, 105-12).


The de-regulated re-emergence of some ephrins and their receptors in adults also has been observed to contribute to tumor invasion, metastasis and neo-angiogenesis (Nakamoto et al. (2002) Microsc Res Tech 59, 58-67; Brantley-Sieders et al., supra). Furthermore, some Eph family members have been found to be over-expressed on tumor cells from a variety of human tumors (Brantley-Sieders et al., supra); Marme (2002) Ann Hematol 81 Suppl 2, S66; Booth et al. (2002) Nat Med 8, 1360-1).


EPH receptor A2 (ephrin type-A receptor 2) is a protein that in humans is encoded by the EPHA2 gene.


EphA2 is upregulated in multiple cancers in man, often correlating with disease progression, metastasis and poor prognosis e.g.: breast (Zelinski et al (2001) Cancer Res. 61, 2301-2306; Zhuang et al (2010) Cancer Res. 70, 299-308; Brantley-Sieders et al (2011) PLoS One 6, e24426), lung (Brannan et al (2009) Cancer Prey Res (Phila) 2, 1039-1049; Kinch et al (2003) Clin Cancer Res. 9, 613-618; Guo et al (2013) J Thorac Oncol. 8, 301-308), gastric (Nakamura et al (2005) Cancer Sci. 96, 42-47; Yuan et al (2009) Dig Dis Sci 54, 2410-2417), pancreatic (Mudali et al (2006) Clin Exp Metastasis 23, 357-365), prostate (Walker-Daniels et al (1999) Prostate 41, 275-280), liver (Yang et al (2009) Hepatol Res. 39, 1169-1177) and glioblastoma (Wykosky et al (2005) Mol Cancer Res. 3, 541-551; Li et al (2010) Tumour Biol. 31, 477-488).


The full role of EphA2 in cancer progression is still not defined although there is evidence for interaction at numerous stages of cancer progression including tumour cell growth, survival, invasion and angiogenesis. Downregulation of EphA2 expression suppresses tumour cancer cell propagation (Binda et al (2012) Cancer Cell 22, 765-780), whilst EphA2 blockade inhibits VEGF induced cell migration (Hess et al (2001) Cancer Res. 61, 3250-3255), sprouting and angiogenesis (Cheng et al (2002) Mol Cancer Res. 1, 2-11; Lin et al (2007) Cancer 109, 332-40) and metastatic progression (Brantley-Sieders et al (2005) FASEB J. 19, 1884-1886).


An antibody drug conjugate to EphA2 has been shown to significantly diminish tumour growth in rat and mouse xenograft models (Jackson et al (2008) Cancer Research 68, 9367-9374) and a similar approach has been tried in man although treatment had to be discontinued for treatment related adverse events (Annunziata et al (2013) Invest New drugs 31, 77-84).


Our earlier pending applications GB1707734.8 filed on 15 May 2017, and GB1721259.8 and GB1721265.5 both filed 19 Dec. 2017, describe bicycle peptide ligands having high binding affinity for EphA2. These applications further describe conjugates of the peptide ligands with therapeutic agents, in particular with cytotoxic agents.


SUMMARY OF THE INVENTION

The present inventors have found that replacement of thioether linkages in looped peptides having affinity for EphA2 by alkylamino linkages results in looped peptide conjugates that display similar affinities to EphA2 as the corresponding conjugates made with all thioether linkages. The replacement of thioether linkages by alkylamino linkages is expected to result in improved solubility and/or improved oxidation stability of the conjugates according to the present invention.


Accordingly, in a first aspect the present invention provides a peptide ligand specific for EphA2 comprising a polypeptide comprising three residues selected from cysteine, L-2,3-diaminopropionic acid (Dap), N-beta-alkyl-L-2,3-diaminopropionic acid (N-AlkDap) and N-beta-haloalkyl-L-2,3-diaminopropionic acid (N-HAlkDap), with the proviso that at least one of said three residues is selected from Dap, N-AlkDap or N-HAlkDap, the said three residues being separated by at least two loop sequences, and a molecular scaffold, the peptide being linked to the scaffold by covalent alkylamino linkages with the Dap or N-AlkDap or N-HAlkDap residues of the polypeptide and by thioether linkages with the cysteine residues of the polypeptide when the said three residues include cysteine, such that two polypeptide loops are formed on the molecular scaffold.


Suitably, the peptide ligand comprises an amino acid sequence selected from:





A1-X1-A2-X2-A3


wherein:


A1, A2, and A3 are independently cysteine, L-2,3-diaminopropionic acid (Dap), N-beta-alkyl-L-2,3-diaminopropionic acid (N-AlkDap), or N-beta-haloalkyl-L-2,3-diaminopropionic acid (N-HAlkDap), provided that at least one of A1, A2, and A3 is Dap, N-AlkDap or N-HAlkDap; and


X1 and X2 represent the amino acid residues between the Cysteine, Dap, N-AlkDap or N-HAlkDap residues, wherein each of X1 and X2 independently is a loop sequence of 4, 5, 6 or 7 amino acid residues.


It can be seen that the derivatives of the invention comprise a peptide loop coupled to a scaffold by at least one alkylamino linkage to Dap or N-AlkDap of N-HAlkDap residues and up to two thioether linkages to cysteine.


The prefix “alkyl” in N-AlkDap and N-HAlkDap refers to an alkyl group having from one to four carbon atoms, preferably methyl. The prefix “halo” is used in this context in its normal sense to signify alkyl groups having one or more, suitably one, fluoro-, chloro-, bromo- or iodo-substituents.


When cysteine is present, the thioether linkage(s) provides an anchor during formation of the cyclic peptides as explained further below. In these embodiments, the thioether linkage is suitably a central linkage of the bicyclic peptide conjugate, i.e. in the peptide sequence two residues forming alkylamino linkages in the peptide are spaced from and located on either side of a cysteine residue forming the thioether linkage. The looped peptide structure is therefore a Bicycle peptide conjugate having a central thioether linkage and two peripheral alkylamino linkages. In alternative embodiments, the thioether linkage is placed at the N-terminus or C-terminus of the peptides, the central linkage and the other terminal linkage being selected from Dap, N-AlkDap or N-HAlkDap.


In embodiments of the invention all three of A1, A2, and A3 may suitably be Dap or N-AlkDap or N-HAlkDap. In these embodiments, the peptide ligands of the invention are suitably Bicycle conjugates having a central alkylamino linkage and two peripheral alkylamino linkages, the peptide forming two loops sharing the central alkylamino linkage. In these embodiments, A1, A2, and A3 are suitably all selected from N-AlkDap or N-HAlkDap, most suitably N-AlkDap, because of favourable reaction kinetics with the alkylated Daps.


In embodiments, the peptide ligand of the present invention additionally comprises one or more modifications selected from: N-terminal and/or C-terminal modifications; replacement of one or more amino acid residues with one or more non-natural amino acid residues (such as replacement of one or more polar amino acid residues with one or more isosteric or isoelectronic amino acids; replacement of one or more hydrophobic amino acid residues with other non-natural isosteric or isoelectronic amino acids); addition of a spacer group; replacement of one or more oxidation sensitive amino acid residues with one or more oxidation resistant amino acid residues; replacement of one or more amino acid residues with an alanine, replacement of one or more L-amino acid residues with one or more D-amino acid residues; N-alkylation of one or more amide bonds within the bicyclic peptide ligand; replacement of one or more peptide bonds with a surrogate bond; peptide backbone length modification; substitution of the hydrogen on the a-carbon of one or more amino acid residues with another chemical group, and post-synthetic bioorthogonal modification of amino acids such as cysteine, lysine, glutamate and tyrosine with suitable amine, thiol, carboxylic acid and phenol-reactive reagents.


Suitably, these embodiments may comprise an N-terminal modification using suitable amino-reactive chemistry, and/or C-terminal modification using suitable carboxy-reactive chemistry. For example, the N-terminal modification may comprise the addition of a molecular spacer group which facilitates the conjugation of effector groups and retention of potency of the bicyclic peptide to its target. The spacer group is suitably an oligopeptide group containing from about 5 to about 30 amino acids, such as an Ala, G-Sar10-A group or bAla-Sar10-A group. Alternatively or additionally, the N-terminal and/or C-terminal modification comprises addition of a cytotoxic agent.


In all of the peptide sequences defined herein, one or more tyrosine residues may be replaced by phenylalanine. This has been found to improve the yield of the bicycle peptide product during base-catalyzed coupling of the peptide to the scaffold molecule.


Suitably, the peptide ligand of the invention is a high affinity binder of the human, mouse and dog EphA2 hemopexin domain. Suitably the binding affinity ki is less than about 500 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, or less than about 10 nM. The binding affinity in the context of this specification is the binding affinity as measured by the methods described below.


Suitably, the peptide ligand of the invention is selective for EphA2, but does not cross-react with EphA1, EphA3 or EphA4. Suitably, the binding affinity ki with each of these ligands is greater than about 500 nM, greater than about 1000 nM, or greater than about 10000 nM.


Suitably, the scaffold comprises a (hetero)aromatic or (hetero)alicyclic moiety. Suitably, the scaffold comprises a tris-substituted (hetero)aromatic or (hetero)alicyclic moiety, for example a tris-methylene substituted (hetero)aromatic or (hetero)alicyclic moiety. The (hetero)aromatic or (hetero)alicyclic moiety is suitably a six-membered ring structure, preferably tris-substituted such that the scaffold has a 3-fold symmetry axis. Thus, in certain preferred embodiments, the scaffold is 1,3,5-tris-methylenebenzene scaffold, for example obtained by reacting the peptide with 1,3,5-tris-(bromomethyl)benzene (TBMB). In other preferred embodiments, the scaffold is a 1,3,5-tris-(acetamido)benzene group, which may be derived by coupling the peptide to 1,3,5-tris-(bromoacetamido)benzene (TBAB) as described further below.


The reactive sites are also suitable for forming thioether linkages with the —SH groups of cysteine in embodiments where the third residue is cysteine. The —SH group of cysteine is highly nucleophilic, and in these embodiments it is expected to react first with the electrophilic centres of the scaffold molecule to anchor the peptide to the scaffold molecule, whereafter the amino groups react with the remaining electrophilic centres of the scaffold molecule to form the looped peptide ligand.


In embodiments, the peptide has protecting groups on nucleophilic groups other than the amino groups and —SH groups (when present) intended for forming the alkylamino linkages.


Suitably, the peptide ligands of the invention may be made by a method that comprises reacting, in a nucleophilic substitution reaction, the peptide as defined herein with a scaffold molecule having three or more leaving groups.


In alternative methods, the compounds of the present invention could be made converting two or more side chain groups of the peptide to leaving groups, followed by reacting the peptide, in a nucleophilic substitution reaction, with a scaffold molecule having two or more amino groups.


The nucleophilic substitution reactions may be performed in the presence of a base, for example where the leaving group is a conventional anionic leaving group. The present inventors have found that the yields of cyclised peptide ligands can be greatly increased by suitable choice of solvent and base for the nucleophilic substitution reaction, and furthermore that the preferred solvent and base are different from the prior art solvent and base combinations that involve only the formation of thioether linkages. In particular, the present inventors have found that improved yields are achieved when using a trialkylamine base, i.e. a base of formula NR1R2R3, wherein R1, R2 and R3 are independently C1-C5alkyl groups, suitably C2-C4 alkyl groups, in particular C2-C3 alkyl groups. Especially suitable bases are triethylamine and diisopropylethylamine (DIPEA). These bases have the property of being only weakly nucleophilic, and it is thought that this property accounts for the fewer side reactions and higher yields observed with these bases. The present inventors have further found that the preferred solvents for the nucleophilic substitution reaction are polar and protic solvents, in particular MeCN/H2O (50:50).


In a further aspect, the present invention provides a drug conjugate comprising the peptide ligand according to the invention conjugated to one or more effector and/or functional groups such as a cytotoxic agent or a metal chelator.


Suitably, the conjugate has the cytotoxic agent linked to the peptide ligand by a cleavable bond, such as a disulphide bond. Suitably, the cytotoxic agent is selected from DMI or MMAE.


In embodiments, the drug conjugate has the following structure:




embedded image


wherein: R1, R2, R3 and R4 represent hydrogen or C1-C6 alkyl groups;


Toxin refers to any suitable cytotoxic agent;


Bicycle represents the looped peptide structure;


n represents an integer selected from 1 to 10; and


m represents an integer selected from 0 to 10.


Suitably, either: R1, R2, R3 and R4 are all H; or R1, R2, R3 are all H and R4=methyl; or R1, R2=methyl and R3, R4=H; or R1, R3=methyl and R2, R4=H; or R1, R2=H and R3, R4=C1-C6 alkyl.


The linker between the toxin and the bicycle peptide may comprise a triazole group formed by click-reaction between an azide-functionalized toxin and an alkyne-functionalized bicycle peptide structure (or vice-versa). In other embodiments, the bicycle peptide may contain an amide linkage formed by reaction between a carboxylate-functionalized toxin and the N-terminal amino group of the bicycle peptide.


The linker between the toxin and the bicycle peptide may comprise a cathepsin-cleavable group to provide selective release of the toxin within the target cells. A suitable cathepsin-cleavable group is valine-citrulline.


The linker between the toxin and the bicycle peptide may comprise one or more spacer groups to provide the desired functionality, e.g. binding affinity or cathepsin cleavability, to the conjugate. A suitable spacer group is para-amino benzyl carbamate (PABC) which may be located intermediate the valine-citrulline group and the toxin moiety. PABC is a so-called self-immolating group that spontaneously breaks away from the toxin after cleavage of the cleavable group.


Thus, in embodiments, the bicycle peptide-drug conjugate may have the following structure made up of Toxin-PABC-cit-val-triazole-Bicycle:




embedded image


In further embodiments, the bicycle peptide-drug conjugate may have the following structure made up of Toxin-PABC-cit-val-dicarboxylate-Bicycle:




embedded image


Wherein (alk) is an alkylene group of formula CnH2n wherein n is from 1 to 10 and may be linear or branched, suitably (alk) is n-propylene or n-butylene.


In another aspect, the invention further provides a kit comprising at least a peptide ligand or conjugate according to the present invention.


In a still further aspect, the present invention provides a composition comprising a peptide ligand or conjugate of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.


Moreover, the present invention provides a method for the treatment of disease using a peptide ligand, conjugate, or a composition according to the present invention. Suitably, the disease is a neoplastic disease, such as cancer.


In a further aspect, the present invention provides a method for the diagnosis, including diagnosis of disease using a peptide ligand, or a composition according to the present invention. Thus in general the binding of an analyte to a peptide ligand may be exploited to displace an agent, which leads to the generation of a signal on displacement. For example, binding of analyte (second target) can displace an enzyme (first target) bound to the peptide ligand providing the basis for a binding assay, especially if the enzyme is held to the peptide ligand through its active site.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a reaction scheme for preparation of thioether-linked bicyclic peptide ligands according to the prior art;



FIG. 2 shows a schematic structure of a reference bicyclic peptide ligand exhibiting specific binding to EphA2.



FIG. 3 shows a schematic structure of a first bicyclic peptide ligand according to the present invention;



FIG. 4 shows a schematic structure of a second bicyclic peptide ligand according to the present invention;



FIG. 5 shows a schematic structure of a third bicyclic peptide ligand according to the present invention.





DETAILED DESCRIPTION OF THE INVENTION

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art, such as in the arts of peptide chemistry, cell culture and phage display, nucleic acid chemistry and biochemistry.


Standard techniques are used for molecular biology, genetic and biochemical methods (see Sambrook et al., Molecular Cloning: A Laboratory Manual, 3rd ed., 2001, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Ausubel et al., Short Protocols in Molecular Biology (1999) 4th ed., John Wiley & Sons, Inc.), which are incorporated herein by reference.


The present invention provides a looped peptide structure as defined in claim 1 comprising two peptide loops subtended between three linkages on the molecular scaffold, the central linkage being common to the two loops. The central linkage may be a thioether linkage formed to a cysteine residue of the peptide, or it is an alkylamino linkage formed to a Dap or N-AlkDap or N-HalkDap residue of the peptide. The two outer linkages are suitably alkylamino linkages formed to Dap or N-AlkDap or N-HalkDap residues of the peptide, or one of the outer linkages may be a thioether linkage formed to a cysteine residue of the peptide.


In one embodiment, the peptide ligands of the invention are fully cross-reactive with murine, dog, cynomolgus and human EphA2. In a yet further embodiment, the peptide ligands of the invention are selective for EphA2, but do not cross-react with EphA1, EphA3 or EphA4.


Suitably the binding affinity ki for EphA2 is less than about 500 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, or less than about 10 nM. Suitably, the binding affinity ki with EphA1, EphA3 and/or EphA4 is greater than about 500 nM, greater than about 1000 nM, or greater than about 10000 nM.


The amino acid sequences of specific peptide ligands according to the present invention are defined in the accompanying claims.


It will be appreciated that modified derivatives of the peptide ligands as defined herein are within the scope of the present invention. Examples of such suitable modified derivatives include one or more modifications selected from: N-terminal and/or C-terminal modifications; replacement of one or more amino acid residues with one or more non-natural amino acid residues (such as replacement of one or more polar amino acid residues with one or more isosteric or isoelectronic amino acids; replacement of one or more non-polar amino acid residues with other non-natural isosteric or isoelectronic amino acids); addition of a spacer group; replacement of one or more oxidation sensitive amino acid residues with one or more oxidation resistant amino acid residues; replacement of one or more amino acid residues with an alanine, replacement of one or more L-amino acid residues with one or more D-amino acid residues; N-alkylation of one or more amide bonds within the bicyclic peptide ligand; replacement of one or more peptide bonds with a surrogate bond; peptide backbone length modification; substitution of the hydrogen on the alpha-carbon of one or more amino acid residues with another chemical group, modification of amino acids such as cysteine, lysine, glutamate/aspartate and tyrosine with suitable amine, thiol, carboxylic acid and phenol-reactive reagents so as to functionalise said amino acids, and introduction or replacement of amino acids that introduce orthogonal reactivities that are suitable for functionalisation, for example azide or alkyn-group bearing amino acids that allow functionalisation with alkyn or azide-bearing moieties, respectively.


In one embodiment, the modified derivative comprises an N-terminal and/or C-terminal modification. In a further embodiment, wherein the modified derivative comprises an N-terminal modification using suitable amino-reactive chemistry, and/or C-terminal modification using suitable carboxy-reactive chemistry. In a further embodiment, said N-terminal or C-terminal modification comprises addition of an effector group, including but not limited to a cytotoxic agent, a radiochelator or a chromophore.


In an embodiment, the N-terminal modification comprises the addition of a molecular spacer group which facilitates the conjugation of effector groups and retention of potency of the bicyclic peptide to its target. The spacer group is suitably an oligopeptide group containing from about 5 to about 30 amino acids, such as an Ala, G-Sar10-A or bAla-Sar10-A group. In one embodiment, the spacer group is selected from bAla-Sar10 -A.


In one embodiment, the modified derivative comprises replacement of one or more amino acid residues with one or more non-natural amino acid residues. In this embodiment, non-natural amino acids may be selected having isosteric/isoelectronic side chains which are neither recognised by degradative proteases nor have any adverse effect upon target potency.


Alternatively, non-natural amino acids may be used having constrained amino acid side chains, such that proteolytic hydrolysis of the nearby peptide bond is conformationally and sterically impeded. In particular, these concern proline analogues, bulky sidechains, C-disubstituted derivatives (for example, aminoisobutyric acid, Aib), and cyclo amino acids, a simple derivative being amino-cyclopropylcarboxylic acid.


In a further embodiment, the non-natural amino acid residuesare selected from: 1-naphthylalanine; 2-naphthylalanine; cyclohexylglycine, phenylglycine; tert-butylglycine; 3,4-dichlorophenylalanine; cyclohexylalanine; and homophenylalanine.


In a yet further embodiment, the non-natural amino acid residues are selected from: 1-naphthylalanine; 2-naphthylalanine; and 3,4-dichlorophenylalanine. These substitutions enhance the affinity compared to the unmodified wildtype sequence.


In a yet further embodiment, the non-natural amino acid residues are selected from: 1-naphthylalanine. This substitution provided the greatest level of enhancement of affinity (greater than 7 fold) compared to wildtype.


In one embodiment, the modified derivative comprises replacement of one or more oxidation sensitive amino acid residues with one or more oxidation resistant amino acid residues. In a further embodiment, the modified derivative comprises replacement of a tryptophan residue with a naphthylalanine or alanine residue. This embodiment provides the advantage of improving the pharmaceutical stability profile of the resultant bicyclic peptide ligand.


In one embodiment, the modified derivative comprises replacement of one or more charged amino acid residues with one or more hydrophobic amino acid residues. In an alternative embodiment, the modified derivative comprises replacement of one or more hydrophobic amino acid residues with one or more charged amino acid residues. The correct balance of charged versus hydrophobic amino acid residues is an important characteristic of the bicyclic peptide ligands. For example, hydrophobic amino acid residues influence the degree of plasma protein binding and thus the concentration of the free available fraction in plasma, while charged amino acid residues (in particular arginine) may influence the interaction of the peptide with the phospholipid membranes on cell surfaces. The two in combination may influence half-life, volume of distribution and exposure of the peptide drug, and can be tailored according to the clinical endpoint. In addition, the correct combination and number of charged versus hydrophobic amino acid residues may reduce irritation at the injection site (if the peptide drug has been administered subcutaneously).


In one embodiment, the modified derivative comprises replacement of one or more L-amino acid residues with one or more D-amino acid residues. This embodiment is believed to increase proteolytic stability by steric hindrance and by a propensity of D-amino acids to stabilise-turn conformations (Tugyi et al (2005) PNAS, 102(2), 413-418).


In all of the peptide sequences defined herein, one or more tyrosine residues may be replaced by phenylalanine. This has been found to improve the yield of the bicycle peptide product during base-catalyzed coupling of the peptide to the scaffold molecule.


In one embodiment, the modified derivative comprises removal of any amino acid residues and substitution with alanines. This embodiment provides the advantage of removing potential proteolytic attack site(s).


It should be noted that each of the above mentioned modifications serve to deliberately improve the potency or stability of the peptide. Further potency improvements based on modifications may be achieved through the following mechanisms:


Incorporating hydrophobic moieties that exploit the hydrophobic effect and lead to lower off rates, such that higher affinities are achieved;


Incorporating charged groups that exploit long-range ionic interactions, leading to faster on rates and to higher affinities (see for example Schreiber et al, Rapid, electrostatically assisted association of proteins (1996), Nature Struct. Biol. 3, 427-31); and


Incorporating additional constraint into the peptide, by for example constraining side chains of amino acids correctly such that loss in entropy is minimal upon target binding, constraining the torsional angles of the backbone such that loss in entropy is minimal upon target binding and introducing additional cyclisations in the molecule for identical reasons.


(for reviews see Gentilucci et al, Curr. Pharmaceutical Design, (2010), 16, 3185-203, and Nestor et al, Curr. Medicinal Chem (2009), 16, 4399-418).


The present invention includes all pharmaceutically acceptable (radio)isotope-labeled compounds of the invention, i.e. compounds of formula (II), wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature, and compounds of formula (I1), wherein metal chelating groups are attached (termed “effector”) that are capable of holding relevant (radio)isotopes, and compounds of formula (1), wherein certain functional groups are covalently replaced with relevant (radio)isotopes or isotopically labelled functional groups.


Examples of isotopes suitable for inclusion in the compounds of the invention comprise isotopes of hydrogen, such as 2H (D) and 3H (T), carbon, such as 11C, 13C and 14C, chlorine, such as 36Cl, fluorine, such as 18F, iodine, such as 123I, 125I and 131I, nitrogen, such as 13N and 15N, oxygen, such as 15O, 17O and 18O, phosphorus, such as 32P, sulfur, such as 35S, copper, such as 64Cu, gallium, such as 67Ga or 68Ga, yttrium, such as 90Y and lutetium, such as 177Lu, and Bismuth, such as 213Bi.


Certain isotopically-labelled compounds of formula (II), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies, and to clinically assess the presence and/or absence of the EphA2 target on diseased tissues such as tumours and elsewhere. The compounds of formula (II) can further have valuable diagnostic properties in that they can be used for detecting or identifying the formation of a complex between a labelled compound and other molecules, peptides, proteins, enzymes or receptors. The detecting or identifying methods can use compounds that are labelled with labelling agents such as radioisotopes, enzymes, fluorescent substances, luminous substances (for example, luminol, luminol derivatives, luciferin, aequorin and luciferase), etc. The radioactive isotopes tritium, i.e. 3H (T), and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.


Substitution with heavier isotopes such as deuterium, i.e. 2H (D), may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.


Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, a N, can be useful in Positron Emission Topography (PET) studies for examining target occupancy.


Incorporation of isotopes into metal chelating effector groups, such as 64Cu, 67Ga, 68Ga, and 177Lu can be useful for visualizing tumour specific antigens employing PET or SPECT imaging.


Incorporation of isotopes into metal chelating effector groups, such as, but not limited to 90Y, 177Lu, and 213Bi, can present the option of targeted radiotherapy, whereby metal-chelator-bearing compounds of formula (II) carry the therapeutic radionuclide towards the target protein and site of action.


Isotopically-labeled compounds of formula (II) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.


Specificity, in the context herein, refers to the ability of a ligand to bind or otherwise interact with its cognate target to the exclusion of entities which are similar to the target. For example, specificity can refer to the ability of a ligand to inhibit the interaction of a human enzyme, but not a homologous enzyme from a different species. Using the approach described herein, specificity can be modulated, that is increased or decreased, so as to make the ligands more or less able to interact with homologues or paralogues of the intended target. Specificity is not intended to be synonymous with activity, affinity or avidity, and the potency of the action of a ligand on its target (such as, for example, binding affinity or level of inhibition) are not necessarily related to its specificity.


Binding activity, as used herein, refers to quantitative binding measurements taken from binding assays, for example as described herein. Therefore, binding activity refers to the amount of peptide ligand which is bound at a given target concentration.


Multispecificity is the ability to bind to two or more targets. Typically, binding peptides are capable of binding to a single target, such as an epitope in the case of an antibody, due to their conformational properties. However, peptides can be developed which can bind to two or more targets; dual specific antibodies, for example, as known in the art as referred to above. In the present invention, the peptide ligands can be capable of binding to two or more targets and are therefore multispecific. Suitably, they bind to two targets, and are dual specific. The binding may be independent, which would mean that the binding sites for the targets on the peptide are not structurally hindered by the binding of one or other of the targets. In this case, both targets can be bound independently. More generally, it is expected that the binding of one target will at least partially impede the binding of the other.


There is a fundamental difference between a dual specific ligand and a ligand with specificity which encompasses two related targets. In the first case, the ligand is specific for both targets individually, and interacts with each in a specific manner. For example, a first loop in the ligand may bind to a first target, and a second loop to a second target. In the second case, the ligand is non-specific because it does not differentiate between the two targets, for example by interacting with an epitope of the targets which is common to both.


In the context of the present invention, it is possible that a ligand which has activity in respect of, for example, a target and an orthologue, could be a bispecific ligand. However, in one embodiment the ligand is not bispecific, but has a less precise specificity such that it binds both the target and one or more orthologues. In general, a ligand which has not been selected against both a target and its orthologue is less likely to be bispecific due to the absence of selective pressure towards bispecificity. The loop length in the bicyclic peptide may be decisive in providing a tailored binding surface such that good target and orthologue cross-reactivity can be obtained, while maintaining high selectivity towards less related homologues.


If the ligands are truly bispecific, in one embodiment at least one of the target specificities of the ligands will be common amongst the ligands selected, and the level of that specificity can be modulated by the methods disclosed herein. Second or further specificities need not be shared, and need not be the subject of the procedures set forth herein.


The peptide ligand compounds of the invention comprise, consist essentially of, or consist of, the peptide covalently bound to a molecular scaffold. The term “scaffold” or “molecular scaffold” herein refers to a chemical moiety that is bonded to the peptide at the alkylamino linkages and thioether linkage (when cysteine is present) in the compounds of the invention. The term “scaffold molecule” or “molecular scaffold molecule” herein refers to a molecule that is capable of being reacted with a peptide or peptide ligand to form the derivatives of the invention having alkylamino and, in certain embodiments, also thioether bonds. Thus, the scaffold molecule has the same structure as the scaffold moiety except that respective reactive groups (such as leaving groups) of the molecule are replaced by alkylamino and thioether bonds to the peptide in the scaffold moiety.


In embodiments, the scaffold is an aromatic molecular scaffold, i.e. a scaffold comprising a (hetero)aryl group. As used herein, “(hetero)aryl” is meant to include aromatic rings, for example, aromatic rings having from 4 to 12 members, such as phenyl rings. These aromatic rings can optionally contain one or more heteroatoms (e.g., one or more of N, O, S, and P), such as thienyl rings, pyridyl rings, and furanyl rings. The aromatic rings can be optionally substituted. “(hetero)aryl” is also meant to include aromatic rings to which are fused one or more other aryl rings or non-aryl rings. For example, naphthyl groups, indole groups, thienothienyl groups, dithienothienyl, and 5,6,7,8-tetrahydro-2-naphthyl groups (each of which can be optionally substituted) are aryl groups for the purposes of the present application. As indicated above, the aryl rings can be optionally substituted. Suitable substituents include alkyl groups (which can optionally be substituted), other aryl groups (which may themselves be substituted), heterocyclic rings (saturated or unsaturated), alkoxy groups (which is meant to include aryloxy groups (e.g., phenoxy groups)), hydroxy groups, aldehyde groups, nitro groups, amine groups (e.g., unsubstituted, or mono- or di-substituted with aryl or alkyl groups), carboxylic acid groups, carboxylic acid derivatives (e.g., carboxylic acid esters, amides, etc.), halogen atoms (e.g., Cl, Br, and I), and the like.


Suitably, the scaffold comprises a tris-substituted (hetero)aromatic or (hetero)alicyclic moiety, for example a tris-methylene substituted (hetero)aromatic or (hetero)alicyclic moiety. The (hetero)aromatic or (hetero)alicyclic moiety is suitably a six-membered ring structure, preferably tris-substituted such that the scaffold has a 3-fold symmetry axis.


In embodiments, the scaffold is a tris-methylene (hetero)aryl moiety, for example a 1,3,5-tris methylene benzene moiety. In these embodiments, the corresponding scaffold molecule suitably has a leaving group on the methylene carbons. The methylene group then forms the R1 moiety of the alkylamino linkage as defined herein. In these methylene-substituted (hetero)aromatic compounds, the electrons of the aromatic ring can stabilize the transition state during nucleophilic substitution. Thus, for example, benzyl halides are 100-1000 times more reactive towards nucleophilic substitution than alkyl halides that are not connected to a (hetero)aromatic group.


In these embodiments, the scaffold and scaffold molecule have the general formula:




embedded image


Where LG represents a leaving group as described further below for the scaffold molecule, or LG (including the adjacent methylene group forming the R1 moiety of the alkylamino group) represents the alkylamino linkage to the peptide in the conjugates of the invention.


In embodiments, the group LG above may be a halogen such as, but not limited to, a bromine atom, in which case the scaffold molecule is 1,3,5-Tris(bromomethyl)benzene (TBMB). Another suitable molecular scaffold molecule is 2,4,6-tris(bromomethyl) mesitylene. It is similar to 1,3,5-tris(bromomethyl) benzene but contains additionally three methyl groups attached to the benzene ring. In the case of this scaffold, the additional methyl groups may form further contacts with the peptide and hence add additional structural constraint. Thus, a different diversity range is achieved than with 1,3,5-Tris(bromomethyl)benzene.


Another preferred molecule for forming the scaffold for reaction with the peptide by nucleophilic substitution is 1,3,5-tris(bromoacetamido)benzene (TBAB):




embedded image


In other embodiments, the scaffold is a non-aromatic molecular scaffold, e.g. a scaffold comprising a (hetero)alicyclic group. As used herein, “(hetero)alicyclic” refers to a homocyclic or heterocyclic saturated ring. The ring can be unsubstituted, or it can be substituted with one or more substituents. The substituents can be saturated or unsaturated, aromatic or nonaromatic, and examples of suitable substituents include those recited above in the discussion relating to substituents on alkyl and aryl groups. Furthermore, two or more ring substituents can combine to form another ring, so that “ring”, as used herein, is meant to include fused ring systems. In these embodiments, the alicyclic scaffold is preferably 1,1′,1″-(1,3,5-triazinane-1,3,5-triyl)triprop-2-en-1-one (TATA).


In other embodiments the molecular scaffold may have a tetrahedral geometry such that reaction of four functional groups of the encoded peptide with the molecular scaffold generates not more than two product isomers. Other geometries are also possible; indeed, an almost infinite number of scaffold geometries is possible, leading to greater possibilities for peptide ligand diversification.


The peptides used to form the ligands of the invention comprise Dap or N-AlkDap or N-HAlkDap residues for forming alkylamino linkages to the scaffold. The structure of diaminopropionic acid is analogous to and isosteric that of cysteine that has been used to form thioether bonds to the scaffold in the prior art, with replacement of the terminal —SH group of cysteine by —NH2:




embedded image


The term “alkylamino” is used herein in its normal chemical sense to denote a linkage consisting of NH or N(R3) bonded to two carbon atoms, wherein the carbon atoms are independently selected from alkyl, alkylene, or aryl carbon atoms and R3 is an alkyl group. Suitably, the alkylamino linkages of the invention comprise an NH moiety bonded to two saturated carbon atoms, most suitably methylene (—CH2—) carbon atoms. The alkylamino linkages of the invention have general formula:





S—R1—N(R3)—R2—P


Wherein:


S represents the scaffold core, e.g. a (hetero)aromatic or (hetero)alicyclic ring as explained further below;


R1 is C1to C3 alkylene groups, suitably methylene or ethylene groups, and most suitably methylene (CH2);


R2 is the methylene group of the Dap or N-AlkDap side chain


R3 is H or C1-4 alkyl including branched alkyl and cycloalkyl, for example methyl, wherein any of the alkyl groups is optionally halogenated; and


P represents the peptide backbone, i.e. the R2 moiety of the above linkage is linked to the carbon atom in the peptide backbone adjacent to a carboxylic carbon of the Dap or N-AlkDap or N-HAlkDap residue.


Certain bicyclic peptide ligands of the present invention have a number of advantageous properties which enable them to be considered as suitable drug-like molecules for injection, inhalation, nasal, ocular, oral or topical administration. Such advantageous properties include:


Species cross-reactivity. This is a typical requirement for preclinical pharmacodynamics and pharmacokinetic evaluation;


Protease stability. Bicyclic peptide ligands should ideally demonstrate stability to plasma proteases, epithelial (“membrane-anchored”) proteases, gastric and intestinal proteases, lung surface proteases, intracellular proteases and the like. Protease stability should be maintained between different species such that a bicycle lead candidate can be developed in animal models as well as administered with confidence to humans;


Desirable solubility profile. This is a function of the proportion of charged and hydrophilic versus hydrophobic residues and intra/inter-molecular H-bonding, which is important for formulation and absorption purposes; and


An optimal plasma half-life in the circulation. Depending upon the clinical indication and treatment regimen, it may be required to develop a bicyclic peptide for short exposure in an acute illness management setting, or develop a bicyclic peptide with enhanced retention in the circulation, and is therefore optimal for the management of more chronic disease states. Other factors driving the desirable plasma half-life are requirements of sustained exposure for maximal therapeutic efficiency versus the accompanying toxicology due to sustained exposure of the agent.


It will be appreciated that salt forms are within the scope of this invention, and references to peptide ligands of the present invention include the salt forms of said compounds.


The salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods such as methods described in Pharmaceutical Salts: Properties, Selection, and Use, P. Heinrich Stahl (Editor), Camille G. Wermuth (Editor), ISBN: 3-90639-026-8, Hardcover, 388 pages, August 2002. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with the appropriate base or acid in water or in an organic solvent, or in a mixture of the two.


Acid addition salts (mono- or di-salts) may be formed with a wide variety of acids, both inorganic and organic. Examples of acid addition salts include mono- or di-salts formed with an acid selected from the group consisting of acetic, 2,2-dichloroacetic, adipic, alginic, ascorbic (e.g. L-ascorbic), L-aspartic, benzenesulfonic, benzoic, 4-acetamidobenzoic, butanoic, (+) camphoric, camphor-sulfonic, (+)-(1S)-camphor-10-sulfonic, capric, caproic, caprylic, cinnamic, citric, cyclamic, dodecylsulfuric, ethane-1,2-disulfonic, ethanesulfonic, 2-hydroxyethanesulfonic, formic, fumaric, galactaric, gentisic, glucoheptonic, D-gluconic, glucuronic (e.g. D-glucuronic), glutamic (e.g. L-glutamic), α-oxoglutaric, glycolic, hippuric, hydrohalic acids (e.g. hydrobromic, hydrochloric, hydriodic), isethionic, lactic (e.g. (+)-L-lactic, (±)-DL-lactic), lactobionic, maleic, malic, (−)-L-malic, malonic, (±)-DL-mandelic, methanesulfonic, naphthalene-2-sulfonic, naphthalene-1,5-disulfonic, 1-hydroxy-2-naphthoic, nicotinic, nitric, oleic, orotic, oxalic, palmitic, pamoic, phosphoric, propionic, pyruvic, L-pyroglutamic, salicylic, 4-amino-salicylic, sebacic, stearic, succinic, sulfuric, tannic, (+)-L-tartaric, thiocyanic, p-toluenesulfonic, undecylenic and valeric acids, as well as acylated amino acids and cation exchange resins.


One particular group of salts consists of salts formed from acetic, hydrochloric, hydroiodic, phosphoric, nitric, sulfuric, citric, lactic, succinic, maleic, malic, isethionic, fumaric, benzenesulfonic, toluenesulfonic, sulfuric, methanesulfonic (mesylate), ethanesulfonic, naphthalenesulfonic, valeric, propanoic, butanoic, malonic, glucuronic and lactobionic acids. One particular salt is the hydrochloride salt. Another particular salt is the acetate salt.


If the compound is anionic, or has a functional group which may be anionic (e.g., —COOH may be —COO), then a salt may be formed with an organic or inorganic base, generating a suitable cation. Examples of suitable inorganic cations include, but are not limited to, alkali metal ions such as Li+, Na+ and K+, alkaline earth metal cations such as Ca2+ and Mg2+, and other cations such as Al3+ or Zn+. Examples of suitable organic cations include, but are not limited to, ammonium ion (i.e., NH4+) and substituted ammonium ions (e.g., NH3R+, NH2R2+, NHR3+, NR4+). Examples of some suitable substituted ammonium ions are those derived from: methylamine, ethylamine, diethylamine, propylamine, dicyclohexylamine, triethylamine, butylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, benzylamine, phenylbenzylamine, choline, meglumine, and tromethamine, as well as amino acids, such as lysine and arginine. An example of a common quaternary ammonium ion is N(CH3)4+.


Where the compounds of the present invention contain an amine function, these may form quaternary ammonium salts, for example by reaction with an alkylating agent according to methods well known to the skilled person. Such quaternary ammonium compounds are within the scope of the invention.


Several conjugated peptides may be incorporated together into the same molecule according to the present invention. For example two such peptide conjugates of the same specificity can be linked together via the molecular scaffold, increasing the avidity of the derivative for its targets. Alternatively, in another embodiment a plurality of peptide conjugates are combined to form a multimer. For example, two different peptide conjugates are combined to create a multispecific molecule. Alternatively, three or more peptide conjugates, which may be the same or different, can be combined to form multispecific derivatives. In one embodiment multivalent complexes may be constructed by linking together the molecular scaffolds, which may be the same or different.


The peptide ligands of the present invention may be made by a method comprising: providing a suitable peptide and a scaffold molecule; and forming the thioether (when cysteine is present) and alkylamino linkages between the peptide and the scaffold molecule.


The peptides for preparation of the peptide ligands of the invention can be made using conventional solid-phase synthesis from amino acid starting materials, which may include appropriate protecting groups as described herein. These methods for making peptides are well known in the art.


Suitably, the peptide has protecting groups on nucleophilic groups other than the —SH and amine groups intended for forming the alkylamino linkages. The nucleophilicity of amino acid side chains has been subject to several studies, and listed in descending order: thiolate in cysteines, amines in Lysine, secondary amine in Histidine and Tryptophan, guanidino amines in Arginine, hydroxyls in Serine/Threonine, and finally carboxylates in aspartate and glutamate. Accordingly, in some cases it may be necessary to apply protecting groups to the more nucleophilic groups on the peptide to prevent undesired side reactions with these groups.


In embodiments, the method comprises: synthesising a peptide having protecting groups on nucleophilic groups other than the amine groups intended for forming the alkylamino linkages and second protecting groups on the amine groups intended for forming alkylamino linkages, wherein the protecting groups on the amine groups intended for forming alkylamino linkages can be removed under conditions different than for the protecting groups on the other nucleophilic groups, followed by treating the peptide under conditions selected to deprotect the amine groups intended for forming alkylamino linkages without deprotecting the other nucleophilic groups. The coupling reaction to the scaffold is then performed, followed by removal of the remaining protecting groups to yield the peptide conjugate.


Suitably, the method comprises reacting, in a nucleophilic substitution reaction, the peptide having the reactive side chain —SH and amine groups, with a scaffold molecule having three or more leaving groups.


The term “leaving group” herein is used in its normal chemical sense to mean a moiety capable of nucleophilic displacement by an amine group. Any such leaving group can be used here provided it is readily removed by nucleophilic displacement by amine. Suitable leaving groups are conjugate bases of acids having a pKa of less than about 5. Non-limiting examples of leaving groups useful in the invention include halo, such as bromo, chloro, iodo, O-tosylate (OTos), O-mesylate (OMes), O-triflate (OTf) or O-trimethylsilyl (OTMS).


The nucleophilic substitution reactions may be performed in the presence of a base, for example where the leaving group is a conventional anionic leaving group. The present inventors have found that the yields of cyclised peptide ligands can be greatly increased by suitable choice of solvent and base (and pH) for the nucleophilic substitution reaction, and furthermore that the preferred solvent and base are different from the prior art solvent and base combinations that involve only the formation of thioether linkages. In particular, the present inventors have found that improved yields are achieved when using a trialkylamine base, i.e. a base of formula NR1R2R3, wherein R1, R2 and R3 are independently C1-C5alkyl groups, suitably C2-C4 alkyl groups, in particular C2-C3 alkyl groups. Especially suitable bases are triethylamine and diisopropylethylamine (DIPEA). These bases have the property of being only weakly nucleophilic, and it is thought that this property accounts for the fewer side reactions and higher yields observed with these bases. The present inventors have further found that the preferred solvents for the nucleophilic substitution reaction are polar and protic solvents, in particular MeCN/H2O containing MeCN and H2O in volumetric ratios from 1:10 to 10:1, suitably from 2:10 to 10:2 and more suitably from 3:10 to 10:3, in particular from 4:10 to 10:4.


Additional binding or functional activities may be attached to the N or C terminus of the peptide covalently linked to a molecular scaffold. The functional group is, for example, selected from the group consisting of: a group capable of binding to a molecule which extends the half-life of the peptide ligand in vivo, and a molecule which extends the half-life of the peptide ligand in vivo. Such a molecule can be, for instance, HSA or a cell matrix protein, and the group capable of binding to a molecule which extends the half-life of the peptide ligand in vivo is an antibody or antibody fragment specific for HSA or a cell matrix protein. Such a molecule may also be a conjugate with high molecular weight PEGs.


In one embodiment, the functional group is a binding molecule, selected from the group consisting of a second peptide ligand comprising a peptide covalently linked to a molecular scaffold, and an antibody or antibody fragment. 2, 3, 4, 5 or more peptide ligands may be joined together. The specificities of any two or more of these derivatives may be the same or different; if they are the same, a multivalent binding structure will be formed, which has increased avidity for the target compared to univalent binding molecules. The molecular scaffolds, moreover, may be the same or different, and may subtend the same or different numbers of loops.


The functional group can moreover be an effector group, for example an antibody Fc region.


Attachments to the N or C terminus may be made prior to binding of the peptide to a molecular scaffold, or afterwards. Thus, the peptide may be produced (synthetically, or by biologically derived expression systems) with an N or C terminal peptide group already in place. Preferably, however, the addition to the N or C terminus takes place after the peptide has been combined with the molecular backbone to form a conjugate. For example, Fluorenylmethyloxycarbonyl chloride can be used to introduce the Fmoc protective group at the N-terminus of the peptide. Fmoc binds to serum albumins including HSA with high affinity, and Fmoc-Trp or Fmoc-Lys bind with an increased affinity. The peptide can be synthesised with the Fmoc protecting group left on, and then coupled with the scaffold through the alkylaminos. An alternative is the palmitoyl moiety which also binds HSA and has, for example been used in Liraglutide to extend the half-life of this GLP-1 analogue.


Alternatively, a conjugate of the peptide with the scaffold can be made, and then modified at the N-terminus, for example with the amine- and sulfhydryl-reactive linker N-e-maleimidocaproyloxy) succinimide ester (EMCS). Via this linker the peptide conjugate can be linked to other peptides, for example an antibody Fc fragment.


The binding function may be another peptide bound to a molecular scaffold, creating a multimer; another binding protein, including an antibody or antibody fragment; or any other desired entity, including serum albumin or an effector group, such as an antibody Fc region.


Additional binding or functional activities can moreover be bound directly to the molecular scaffold.


In embodiments, the scaffold may further comprise a reactive group to which the additional activities can be bound. Preferably, this group is orthogonal with respect to the other reactive groups on the molecular scaffold, to avoid interaction with the peptide. In one embodiment, the reactive group may be protected, and deprotected when necessary to conjugate the additional activities.


Accordingly, in a further aspect of the invention, there is provided a drug conjugate comprising a peptide ligand as defined herein conjugated to one or more effector and/or functional groups.


Effector and/or functional groups can be attached, for example, to the N or C termini of the polypeptide, or to the molecular scaffold.


Appropriate effector groups include antibodies and parts or fragments thereof. For instance, an effector group can include an antibody light chain constant region (CL), an antibody CH1 heavy chain domain, an antibody CH2 heavy chain domain, an antibody CH3 heavy chain domain, or any combination thereof, in addition to the one or more constant region domains. An effector group may also comprise a hinge region of an antibody (such a region normally being found between the CH1 and CH2 domains of an IgG molecule).


In a further embodiment of this aspect of the invention, an effector group according to the present invention is an Fc region of an IgG molecule. Advantageously, a peptide ligand-effector group according to the present invention comprises or consists of a peptide ligand Fc fusion having a tβ half-life of a day or more, two days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more or 7 days or more. Most advantageously, the peptide ligand according to the present invention comprises or consists of a peptide ligand Fc fusion having a tβ half-life of a day or more.


Functional groups include, in general, binding groups, drugs, reactive groups for the attachment of other entities, functional groups which aid uptake of the macrocyclic peptides into cells, and the like.


The ability of peptides to penetrate into cells will allow peptides against intracellular targets to be effective. Targets that can be accessed by peptides with the ability to penetrate into cells include transcription factors, intracellular signalling molecules such as tyrosine kinases and molecules involved in the apoptotic pathway. Functional groups which enable the penetration of cells include peptides or chemical groups which have been added either to the peptide or the molecular scaffold. Peptides such as those derived from such as VP22, HIV-Tat, a homeobox protein of Drosophila (Antennapedia), e.g. as described in Chen and Harrison, Biochemical Society Transactions (2007) Volume 35, part 4, p821; Gupta et al. in Advanced Drug Discovery Reviews (2004) Volume 57 9637. Examples of short peptides which have been shown to be efficient at translocation through plasma membranes include the 16 amino acid penetratin peptide from Drosophila Antennapedia protein (Derossi et al (1994) J Biol. Chem. Volume 269 p10444), the 18 amino acid ‘model amphipathic peptide’ (Oehlke et al (1998) Biochim Biophys Acts Volume 1414 p127) and arginine rich regions of the HIV TAT protein. Non peptidic approaches include the use of small molecule mimics or SMOCs that can be easily attached to biomolecules (Okuyama et al (2007) Nature Methods Volume 4 p153). Other chemical strategies to add guanidinium groups to molecules also enhance cell penetration (Elson-Scwab et al (2007) J Biol Chem Volume 282 p13585). Small molecular weight molecules such as steroids may be added to the molecular scaffold to enhance uptake into cells.


One class of functional groups which may be attached to peptide ligands includes antibodies and binding fragments thereof, such as Fab, Fv or single domain fragments. In particular, antibodies which bind to proteins capable of increasing the half-life of the peptide ligand in vivo may be used.


RGD peptides, which bind to integrins which are present on many cells, may also be incorporated.


In one embodiment, a peptide ligand-effector group according to the invention has a tβ half-life selected from the group consisting of: 12 hours or more, 24 hours or more, 2 days or more, 3 days or more, 4 days or more, 5 days or more, 6 days or more, 7 days or more, 8 days or more, 9 days or more, 10 days or more, 11 days or more, 12 days or more, 13 days or more, 14 days or more, 15 days or more or 20 days or more. Advantageously a peptide ligand-effector group or composition according to the invention will have a tβ half life in the range 12 to 60 hours. In a further embodiment, it will have a tβ half-life of a day or more. In a further embodiment still, it will be in the range 12 to 26 hours.


In one particular embodiment of the invention, the functional group conjugated to the looped peptide is selected from a metal chelator, which is suitable for complexing metal radioisotopes of medicinal relevance. Such effectors, when complexed with said radioisotopes, can present useful agents for cancer therapy. Suitable examples include DOTA, NOTA, EDTA, DTPA, HEHA, SarAr and others (Targeted Radionuclide therapy, Tod Speer, Wolters/Kluver Lippincott Williams & Wilkins, 2011).


Possible effector groups also include enzymes, for instance such as carboxypeptidase G2 for use in enzyme/prodrug therapy, where the peptide ligand replaces antibodies in ADEPT.


In one particular embodiment of this aspect of the invention, the functional group is selected from a drug, such as a cytotoxic agent for cancer therapy. Suitable examples include: alkylating agents such as cisplatin and carboplatin, as well as oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, ifosfamide; Anti-metabolites including purine analogs azathioprine and mercaptopurine or pyrimidine analogs; plant alkaloids and terpenoids including vinca alkaloids such as Vincristine, Vinblastine, Vinorelbine and Vindesine; Podophyllotoxin and its derivatives etoposide and teniposide; Taxanes, including paclitaxel, originally known as Taxol; topoisomerase inhibitors including camptothecins: irinotecan and topotecan, and type II inhibitors including amsacrine, etoposide, etoposide phosphate, and teniposide. Further agents can include antitumour antibiotics which include the immunosuppressant dactinomycin (which is used in kidney transplantations), doxorubicin, epirubicin, bleomycin and others.


In one further particular embodiment of the invention according to this aspect, the cytotoxic agent is selected from DM1 or MMAE.


DM1 is a cytotoxic agent which is a thiol-containing derivative of maytansine and has the following structure:




embedded image


Monomethyl auristatin E (MMAE) is a synthetic antineoplastic agent and has the following structure:




embedded image


In one embodiment, the cytotoxic agent is linked to the bicyclic peptide by a cleavable bond, such as a disulphide bond. In a further embodiment, the groups adjacent to the disulphide bond are modified to control the hindrance of the disulphide bond, and by this the rate of cleavage and concomitant release of cytotoxic agent.


Published work established the potential for modifying the susceptibility of the disulphide bond to reduction by introducing steric hindrance on either side of the disulphide bond (Kellogg et al (2011) Bioconjugate Chemistry, 22, 717). A greater degree of steric hindrance reduces the rate of reduction by intracellular glutathione and also extracellular (systemic) reducing agents, consequentially reducing the ease by which toxin is released, both inside and outside the cell. Thus, selection of the optimum in disulphide stability in the circulation (which minimises undesirable side effects of the toxin) versus efficient release in the intracellular milieu (which maximises the therapeutic effect) can be achieved by careful selection of the degree of hindrance on either side of the disulphide bond.


The hindrance on either side of the disulphide bond is modulated through introducing one or more methyl groups on either the targeting entity (here, the bicyclic peptide) or toxin side of the molecular construct.


Thus, in one embodiment, the cytotoxic agent is selected from a compound of formula:




embedded image


wherein n represents an integer selected from 1 to 10; and


R1 and R2 independently represent hydrogen or methyl groups.


In one embodiment of the compound of the above formula, n represents 1 and R1 and R2 both represent hydrogen (i.e. the maytansine derivative DM1).


In an alternative embodiment of the compound of the above formula, n represents 2, R1 represents hydrogen and R2 represents a methyl group (i.e. the maytansine derivative DM3).


In one embodiment of the compound, n represents 2 and R1 and R2 both represent methyl groups (i.e. the maytansine derivative DM4).


It will be appreciated that the cytotoxic agent can form a disulphide bond, and in a conjugate structure with a bicyclic peptide, the disulphide connectivity between the thiol-toxin and thiol-bicycle peptide is introduced through several possible synthetic schemes.


In one embodiment, the bicyclic peptide component of the conjugate has the following structure:




embedded image


wherein m represents an integer selected from 0 to 10,


Bicycle represents any suitable looped peptide structure as described herein; and


R3 and R4 independently represent hydrogen or methyl.


Compounds of the above formula where R3 and R4 are both hydrogen are considered unhindered and compounds of the above formula where one or all of R3 and R4 represent methyl are considered hindered.


It will be appreciated that the bicyclic peptide of the above formula can form a disulphide bond, and in a conjugate structure with a cytotoxic agent described above, the disulphide connectivity between the thiol-toxin and thiol-bicycle peptide is introduced through several possible synthetic schemes.


In one embodiment, the cytotoxic agent is linked to the bicyclic peptide by the following linker:




embedded image


wherein R1, R2, R3 and R4 represent hydrogen or C1-C6 alkyl groups;


Toxin refers to any suitable cytotoxic agent defined herein;


Bicycle represents any suitable looped peptide structure as described herein;


n represents an integer selected from 1 to 10; and


m represents an integer selected from 0 to 10.


When R1, R2, R3 and R4 are each hydrogen, the disulphide bond is least hindered and most susceptible to reduction. When R1, R2, R3 and R4 are each alkyl, the disulphide bond is most hindered and least susceptible to reduction. Partial substitutions of hydrogen and alkyl yield a gradual increase in resistance to reduction, and concomitant cleavage and release of toxin. Preferred embodiments include: R1, R2, R3 and R4 all H; R1, R2, R3 all H and R4=methyl; R1, R2=methyl and R3, R4=H; R1, R3=methyl and R2, R4=H; and R1, R2=H, R3, R4=C1-C6 alkyl.


In one embodiment, the toxin of compound is a maytansine and the conjugate comprises a compound of the following formula:




embedded image


wherein R1, R2, R3 and R4 are as defined above;


Bicycle represents any suitable looped peptide structure as defined herein;


n represents an integer selected from 1 to 10; and


m represents an integer selected from 0 to 10.


Further details and methods of preparing the above-described conjugates of bicycle peptide ligands with toxins are described in detail in our published patent applications WO2016/067035 and WO2017/191460. The entire disclosure of these applications is expressly incorporated herein by reference.


The linker between the toxin and the bicycle peptide may comprise a triazole group formed by click-reaction between an azide-functionalized toxin and an alkyne-functionalized bicycle peptide structure (or vice-versa). In other embodiments, the bicycle peptide may contain an amide linkage formed by reaction between a carboxylate-functionalized toxin and the N-terminal amino group of the bicycle peptide.


The linker between the toxin and the bicycle peptide may comprise a cathepsin-cleavable group to provide selective release of the toxin within the target cells. A suitable cathepsin-cleavable group is valine-citrulline.


The linker between the toxin and the bicycle peptide may comprise one or more spacer groups to provide the desired functionality, e.g. binding affinity or cathepsin cleavability, to the conjugate. A suitable spacer group is para-amino benzyl carbamate (PABC) which may be located intermediate the valine-citrulline group and the toxin moiety.


Thus, in embodiments, the bicycle peptide-drug conjugate may have the following structure made up of Toxin-PABC-cit-val-triazole-Bicycle:




embedded image


In further embodiments, the bicycle peptide-drug conjugate may have the following structure made up of Toxin-PABC-cit-val-dicarboxylate-Bicycle:




embedded image


wherein (alk) is an alkylene group of formula CnH2n wherein n is from 1 to 10 and may be linear or branched, suitably (alk) is n-propylene or n-butylene.


A detailed description of methods for the preparation of peptide ligand-drug conjugates according to the present invention is given in our earlier applications WO2016/067035 and PCT/EP2017/083954 filed 20th Dec. 2017, the entire contents of which are incorporated herein by reference.


Peptide ligands according to the present invention may be employed in in vivo therapeutic and prophylactic applications, in vitro and in vivo diagnostic applications, in vitro assay and reagent applications, and the like.


In general, the use of a peptide ligand can replace that of an antibody. Derivatives selected according to the invention are of use diagnostically in Western analysis and in situ protein detection by standard immunohistochemical procedures; for use in these applications, the derivatives of a selected repertoire may be labelled in accordance with techniques known in the art. In addition, such peptide ligands may be used preparatively in affinity chromatography procedures, when complexed to a chromatographic support, such as a resin. All such techniques are well known to one of skill in the art. Peptide ligands according to the present invention possess binding capabilities similar to those of antibodies, and may replace antibodies in such assays.


Diagnostic uses include any uses which to which antibodies are normally put, including test-strip assays, laboratory assays and immunodiagnostic assays.


Therapeutic and prophylactic uses of peptide ligands prepared according to the invention involve the administration of derivatives selected according to the invention to a recipient mammal, such as a human. Substantially pure peptide ligands of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human. Once purified, partially or to homogeneity as desired, the selected peptides may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, Academic Press, NY).


Generally, the present peptide ligands will be utilised in purified form together with pharmacologically appropriate carriers. Typically, these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable adjuvants, if necessary to keep a peptide complex in suspension, may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.


Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition).


The peptide ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include antibodies, antibody fragments and various immunotherapeutic drugs, such as cyclosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. Pharmaceutical compositions can include “cocktails” of various cytotoxic or other agents in conjunction with the selected antibodies, receptors or binding proteins thereof of the present invention, or even combinations of selected peptides according to the present invention having different specificities, such as peptides selected using different target derivatives, whether or not they are pooled prior to administration.


The route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art. For therapy, including without limitation immunotherapy, the selected antibodies, receptors or binding proteins thereof of the invention can be administered to any patient in accordance with standard techniques. The administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter. The dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counter-indications and other parameters to be taken into account by the clinician.


The peptide ligands of this invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of activity loss and that use levels may have to be adjusted upward to compensate.


The compositions containing the present peptide ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a “therapeutically-effective dose”. Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of selected peptide ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present peptide ligands or cocktails thereof may also be administered in similar or slightly lower dosages.


A composition containing a peptide ligand according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal. In addition, the selected repertoires of peptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells. Blood from a mammal may be combined extracorporeally with the selected peptide ligands whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.


The invention is further described with reference to the following examples.


EXAMPLES

Materials and Methods


















Precursor
Precursor



Abbreviations
Name
Name
CAS
Supplier







1Nal
1-Naphthylalanine
Fmoc-3-(1-naphthyl)-L-
96402-
Fluorochem




alanine
49-2



2FuAla
2-Furylalanine
Fmoc-L-2-furylalanine
159611-
Combi





02-6
Blocks


2Nal
2-Naphthylalanine
Fmoc-3-(2-naphthyl)-L-
112883-
Alfa Aesar




alanine
43-9



3,3-DPA
3,3-
fmoc-3,3-
189937-
Alfa Aesar



Diphenylalanine
diphenylalanine
46-0



3,4-DCPhe
3,4-
Fmoc-3,4-dichloro-L-
17766-
PolyPeptide



Dichlorophenylalanine
phenylalanine
59-5



3Pal
3-(3-Pyridyl)-
N-Fmoc-3-(3-pyridyl)-
175453-
Fluorochem



Alanine
Lβnine
07-3



4,4-BPA
4,4′-
Fmoc-L-4, 4'-
199110-
Alfa Aesar



Biphenylalanine
Biphenylalanine
64-0



4BenzylPro
4-Benzyl-
Fmoc-4-Benzyl-

PolyPeptide



pyrrolidine-2-
pyrrolidine-2-carboxylic





carboxylic acid
acid




4BrPhe
4-
Fmoc-4-Bromo-L-
198561-
PolyPeptide



Bromophenylalanine
phenylalanine
04-5



4FlPro
4-Fluoro-
Fmoc-4-fluoro-
203866-
PolyPeptide



pyrrolidine-2-
pyrrolidine-2-carboxylic
19-7




carboxylic acid
acid




4MeoPhe
4-
Fmoc-4-
77128-
Iris



Methoxyphenylalanine
Methoxyphenylalanine
72-4
Biotech


4Pal
3-(4-Pyridyl)-
N-Fmoc-3-(4-pyridyl)-
169555-
Fluorochem



Alanine
L-alanine
95-7



4PhenylPro
4-Phenyl-
Fmoc-4-phenyl-
269078-
Cambridge



pyrrolidine-2-
pyrrolidine-2-carboxylic
71-9
Bioscience



carboxylic acid
acid




Ac
Acetyl





AC3C
1-
1-(Fmoc-
126705-
Iris



Aminocyclopropane-
amino)cyclopropane-
22-4
Biotech



l-carboxylic acid
carboxylic acid




AC4C
1-Amino-1-
1-(Fmoc-amino)-
885951-
Fluorochem



cyclobutanecarboxylic
cyclobutylcarboxylic
77-9




acid
acid




AC5C
1-Amino-1-
1-(Fmoc-amino)
117322-
Iris



cyclopentanecarboxylic
cyclopentanecarboxylic
30-2
Biotech



acid
acid




AF488
AlexaF1uor488
AlexaF1uor488-NHS

Fisher




Ester

Scientific


Aib
2-
Fmoc-α-
94744-
Fluorochem



Aminoisobutyric
aminoisobutyric
50-0




acid
acid




Aza-Gly
Azaglycine





Aze
Azetidine
Fmoc-L-azetidine-2-
136552-
Combi




carboxylic acid
06-2
Blocks


β-Ala
β-Alanine
Fmoc-β-alanine
35737-
Fluorochem





10-1



C5g
Cyclopentylglycine
Fmoc-L-
220497-
Fluorochem




cyclopentylglycine
61-0



Cba
β-
Fmoc-β-cyclobutyl-L-
478183-
IRIS



Cyclobutylalanine
alanine
62-9
Biotech






GmbH


Cpa
β-
Fmoc-β-cyclopropyl-L-
214750-
Fluorochem



Cyclopropylalanine
alanine
76-2



Cpg
Cyclopropylglycine
Fmoc-L-
121225
Apollo




cycloproprylglycine
7-18-5
Scientific










DOTA
1,4,7,10-tetraazacyclododecane-1,4,7,10-





tetraacetic acid













Fl
5(6)-


Sigma



carboxyfluorescein





HArg
HomoArginine
Fmoc-L-
401915-
Fluorochem




HomoArg(Pbf)-OH
53-5



HPhe
HomoPhenylalanine
Fmoc-L-
132684-
Iris




Homophenylalanine
59-4
Biotech


HyP
Hydroxyproline
Fmoc-
122996-
Sigma




Hydroxyproline(tBu)-
47-8





OH




NO2Phe
4-
Fmoc-4-nitro-L-
95753-
PolyPeptide



Nitrophenylalanine
phenylalanine
55-2



Phg
Phenylglycine
Fmoc-L-phenylglycine
102410-
Combi





65-1
Blocks


Pip
Pipecolic acid
Fmoc-L-Pipecolic acid
86069-
Peptech





86-5



Sar
Sarcosine, such that Sarx
Fmoc-Sarcosine-OH
77128-
Sigma



represents x Sar

70-2




residues





tBuGly
Tert-leucine
Fmoc-L-tert-leucine
132684-
Fluorochem





60-7



Thi
2-Thienylalanine
Fmoc-2-Thienylalanine
130309-
Novabiochem





35-2



ThiAz
3-(1,2,4-triazol-1-
Fmoc-3-(1,2,4-triazol-1-
121744
Sigma



yl)-Alanine
yl)-Ala-OH
9-37-0



ΨAla
Reduced amide on






backbone









In addition, the following non-natural amino acid precursors were used for the preparation of the DAP and N-MeDAP modified peptides:















Compound
CAS
Mw
Supplier







Fmoc-L-
446847-
440.49
Iris Biotech GMBH


Dap(Boc,Me)-OH
80-9




Fmoc-Dap(Boc)-OH
162558-
426.46
Sigma Aldrich



25-0









Peptide Synthesis


Peptide synthesis was based on Fmoc chemistry, using a Symphony and SymphonyX peptide synthesiser manufactured by Peptide Instruments and a Syro II synthesiser by MultiSynTech. Standard Fmoc-amino acids were employed (Sigma, Merck), with appropriate side chain protecting groups: where applicable standard coupling conditions were used in each case, followed by deprotection using standard methodology. Peptides were purified by HPLC and following isolation they were modified with 1,3,5-tris(bromomethyl)benzene (TBMB, Sigma). For this, linear peptide was diluted with H2O up to ˜35 mL, ˜500 μL of 100 mM TBMB in acetonitrile was added, and the reaction was initiated with ˜5 mL of 1 M NH4HCO3 in H2O. The reaction was allowed to proceed for ˜30-60 min at RT, and quenched with ˜500 ul of the 1M Cysteine hydrochloride (Sigma) once the reaction had completed (judged by MALDI). Following lyophilisation, the modified peptide was purified in a Gemini C18 column (Phenomenex) using water/acetonitrile with 0.1% trifluoroacetic acid as mobile phase. Pure fractions containing the correct cyclised material were pooled, lyophilised and kept at −20° C. for storage.


All amino acids, unless noted otherwise, were used in the L-configurations.


Biological Data


1. Fluorescence polarisation measurements


(a) Direct Binding Assay


Peptides with a fluorescent tag (either fluorescein, SIGMA or Alexa Fluor488™, Fisher Scientific) were diluted to 2.5 nM in PBS with 0.01% tween 20 or 50 mM HEPES with 100 mM NaCl and 0.01% tween pH 7.4 (both referred to as assay buffer). This was combined with a titration of protein in the same assay buffer as the peptide to give 1 nM peptide in a total volume of 25 μL in a black walled and bottomed low bind low volume 384 well plates, typically 54 μL assay buffer, 10 μL protein (Table 1) then 10 μL fluorescent peptide. One in two serial dilutions were used to give 12 different concentrations with top concentrations ranging from 500 nM for known high affinity binders to 10 μM for low affinity binders and selectivity assays. Measurements were conducted on a BMG PHERAstar FS equipped with an “FP 485 520 520” optic module which excites at 485 nm and detects parallel and perpendicular emission at 520 nm. The PHERAstar FS was set at 25° C. with 200 flashes per well and a positioning delay of 0.1 second, with each well measured at 5 to 10 minute intervals for 60 minutes. The gain used for analysis was determined for each tracer at the end of the 60 minutes where there was no protein in the well. Data was analysed using Systat Sigmaplot version 12.0. mP values were fit to a user defined quadratic equation to generate a Kd value: f=ymin+(ymax−ymin)/Lig*((x+Lig+Kd)/2-sqrt((((x+Lig+Kd)/2)^2)−(Lig*x))). “Lig” was a defined value of the concentration of tracer used.


(b) Competition Binding Assay


Peptides without a fluorescent tag were tested in competition with a peptide with a fluorescent tag and a known Kd (Table 2). Peptides were diluted to an appropriate concentration in assay buffer as described in the direct binding assay with a maximum of 5% DMSO, then serially diluted 1 in 2. Five μL of diluted peptide was added to the plate followed by 10 μL of human or mouse EphA2 (Table 1) at a fixed concentration which was dependent on the fluorescent peptide used (Table 2), then 10 μL fluorescent peptide added. Measurements were conducted as for the direct binding assay, however the gain was determined prior to the first measurement. Data analysis was in Systat Sigmaplot version 12.0 where the mP values were fit to a user defined cubic equation to generate a Ki value:






f
=


y





min

+


(


y





max

-

y





min


)

/


Lig
*

(

(



Lig
*



(

(



2
*



(




(

Klig
+
Kcomp
+
Lig
+
Comp
+


Prot
*


c


)




2

-


3
*




(



Kcomp
*



(

Lig
-


Prot
*


c


)


+


Klig
*



(


(

Comp
-


Prot
*


c


)

+


Klig
*


Kcomp


)



)





0.5
*



COS


(


ARCCOS


(


(



-

2
*





(

Klig
+
Kcomp
+
Lig
+
comp
-


Prot
*


c


)




3

+


9
*




(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)

*



(



Kcomp
*



(

Lig
-


Prot
*


c


)


+


Klig
*

(





Comp
-


Prot
*


c


)

+


Klig
*


Kcomp


)


-


27
*



(


-

1
*




Klig
*



Kcomp
*



Prot
*


c

)



)

/

(


2
*



(



(



(




(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)




2

-


3
*



(



Kcomp
*



(

Lig
-


Prot
*


c


)


+





Klig
*






(

Comp
-


Prot
*


c


)

+


Klig
*


Kcomp


)



)




3

)




0.5

)


)


)


/
3

)




)


-

(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)


)

)


/

(


(


3
*


Klig

)

+

(

(



2
*




(




(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)




2

-


3
*



(



Kcomp
*



(

Lig
-


Prot
*


c


)


+


Klig
*



(

Comp
-


Prot
*


c


)


+


Klig
*


Kcomp


)



)





0.5
*



COS


(

ARCCOS


(



(



-

2
*





(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)




3

+


9
*




(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)

*



(



Kcomp
*



(

Lig
-


Prot
*


c


)


+


Klig
*



(

Comp
-


Prot
*


c


)


+


Klig
*


Kcomp


)


-


27
*



(


-

1
*




Klig
*



Kcomp
*



Prot
*


c

)



)

/

(


2
*



(

(



(




(

Klig
+
Kcomp
+
Lig
+


CompProt
*


c


)




2

-


3
*



(



Kcomp
*



(

Lig
-


Prot
*


c


)


+


Klig
*



(

Comp
-


Prot
*


c


)


+


Klig
*


Kcomp


)



0



3


)




0.5

)

)


)


/
3

)


)



-

(

Klig
+
Kcomp
+
Lig
+
Comp
-


Prot
*


c


)


)

)


)










“Lig”, “KLig” and “Prot” were all defined values relating to: fluorescent peptide concentration, the Kd of the fluorescent peptide and EphA2 concentration respectively.









TABLE 1







Ephrin receptors and source















Catalogue


Receptor (domain)
Species
Format/tag
Supplier
number





EphA1 (Ecto)
Human
Fc fusion
R&D systems
7146-A1


EphA2 (Ecto)
Human
C-terminal
R&D systems
3035-A2




polyHis




EphA2 (Ecto)
Human
C-terminal
In-house
N/A




polyHis




EphA2 (Ecto)
Mouse
Fc fusion
R&D systems
639-A2


EphA2 (Ecto)
Mouse
C-terminal
Sino
50586-




polyHis
Biological
M08H


EphA2 (ligand
Rat
C-terminal
In-house
N/A


binding)

polyHis




EphA2 (ligand
Dog
C-terminal
In-house
N/A


binding)

polyHis




EphA3 (Ecto)
Human
Fc fusion
R&D systems
6444-A3


EphA3 (Ecto)
Human
N-terminal
In-house
N/A




polyHis




EphA3 (Ecto)
Rat
C-terminal
Sino
80465-




polyHis
Biological
R08H


EphA4 (Ecto)
Human
Fc fusion
R&D systems
6827-A4


EphA4 (Ecto)
Human
C-terminal
Sino
11314-




polyHis
Biological
H08H


EphA4 (Ecto)
Rat
C-terminal
Sino
80123-




polyHis
Biological
R08H


EphA6 (Ecto)
Human
Fc fusion
R&D systems
5606-A6


EphA7 (Ecto)
Human
Fc fusion
R&D systems
6756-A7


EphB1 (Ecto)
Rat
Fc fusion
R&D systems
1596-B1


EphB4 (Ecto)
human
C-terminal
R&D systems
3038-B4




polyHis
















TABLE 2







Final concentrations of fluorescent peptide and EphA2 as


used with Competition Binding Assays











Concentration
Concentration
Concentration



of fluorescent
of Human
of Mouse


Fluorescent
peptide
EphA2
EphA2


peptide
(nM)
(nM)
(nM)













Compound 1
1
300



Compound 12
10
75



Compound 66
1
30



Compound 18
0.8 (human)
2.4
50



1 (mouse)









The peptide ligands described herein were tested in the above mentioned assays.


Reference Example 1

A first reference Bicyclic Peptide chosen for comparison of thioether to alkylamino scaffold linkage was designated 55-03-05-N233. It is a bicycle conjugate of a thioether-forming peptide with a trimethylene benzene scaffold. The structure of this bicycle derivative is shown schematically in FIG. 2. The linear peptide before conjugation has sequence:


[B-Ala][Sar]10H[dD]VPCPWGPFWCPVNRPGC


Conjugation to 1,3,5-tris(bromomethyl)benzene (TBMB, Sigma) was carried out as follows. The linear peptide was diluted with H2O up to ˜35 mL, ˜500 μL of 100 mM TBMB in acetonitrile was added, and the reaction was initiated with 5 mL of 1 M NH4HCO3 in H2O. The reaction was allowed to proceed for ˜30-60 min at RT, and lyophilised once the reaction had completed (judged by MALDI). Following lyophilisation, the modified peptide was purified with a Gemini C18 column (Phenomenex), and changing the acid to 0.1% trifluoroacetic acid. Pure fractions containing the correct TMB-modified material were pooled, lyophilised and kept at −20° C. for storage.


The resulting Bicycle derivative designated 55-03-05-N233 showed high affinity to EphA2. The measured affinity (Ki) to EphA2 of the derivative was 4.12 nM.


Example 1

A bicycle peptide designated 55-03-05-N314 was made corresponding to the bicycle region of the peptide ligand of Reference Example 1, with replacement of the first and second cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold. The structure of this derivative is shown schematically in FIG. 3.


The linear peptide used to form this bicycle was as follows:





[Ac][B-Ala][Sar]10H[dD]VP[Dap]PWGPFW[Dap]PVNRPGC


Cyclisation with TBMB was performed in a mixture of Acetonitrile/water in the presence of DIPEA as the base for 1-16 hours, as described in more detail in PCT/EP2017/083953 and PCT/EP2017/083954 filed 20 Dec. 2017. Unlike the cyclisation of Reference Example 1, the yield is relatively low when using the conventional NaHCO3 as the base.


The measured Ki with EphA2 was 135.5 nM, which demonstrates that the change to alkylamino linkages in this example resulted in relatively little change in binding affinity relative to the thioether linked derivative of Reference Example 1.


Example 2

A bicycle peptide designated 55-03-05-N316 was made corresponding to the bicycle region of the peptide ligand of Reference Example 1, with replacement of the second and third cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold. The structure of this derivative is shown schematically in FIG. 3.


The linear peptide used to form this bicycle was as follows:





[Ac][B-Ala][Sar]10H[dD]VPCPWGPFW[Dap]PVNRPG[Dap]


Cyclisation with TBMB was performed as described in Example 1.


The measured Ki with EphA2 was 604 nM, which demonstrates that the change to alkylamino linkages in this example preserved a relatively high level of binding affinity relative to the thioether-linked derivative of Reference Example 1.


Example 3

A bicycle peptide designated 55-03-05-N318 was made corresponding to the bicycle region of the peptide ligand of Reference Example 1, with replacement of the first and third cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold. The structure of this derivative is shown schematically in FIG. 4.


The linear peptide used to form this bicycle was as follows:





[Ac][B-Ala][Sar]10H[dD]VP[Dap]PWGPFWCPVNRPG[Dap]


Cyclisation with TBMB was performed as described in Example 1.


The measured Ki with EphA2 was 31.5 nM, which demonstrates that the change to alkylamino linkages in this example resulted in only a minimal change in binding affinity relative to the thioether-linked derivative of Reference Example 1.


Reference Example 2

A first reference Bicyclic Peptide chosen for comparison of thioether to alkylamino scaffold linkage was designated 55-03-05-N238. It is a bicycle conjugate of a thioether-forming peptide with a trimethylene benzene scaffold. The linear peptide before conjugation has sequence:





[B-Ala][Sar]10H[dD]VPC[Aib][1Nal]G[Aib]F[1Nal]CP[tBuGly]N[HArg]P[dD]C


Conjugation to 1,3,5-tris(bromomethyl)benzene (TBMB, Sigma) was carried out as described in Example 1.


The resulting Bicycle derivative designated 55-03-05-N238 showed high affinity to EphA2. The measured affinity (Ki) to EphA2 of the derivative was 19.7 nM.


Example 4

A bicycle peptide designated 55-03-05-N315 was made corresponding to the bicycle region of the peptide ligand of Reference Example 2, with replacement of the first and second cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold.


The linear peptide used to form this bicycle was as follows:





[B-Ala][Sar]10H[dD]VP [Dap][Aib][1Nal]G[Aib]F[1Nal][Dap]P[tBuGly]N[HArg]P[dD]C


Cyclisation with TBMB was performed as described in Example 1.


The measured Ki with EphA2 was 640 nM, which demonstrates that the change to alkylamino linkages in this example preserved a significant binding affinity relative to the thioether-linked derivative of Reference Example 2.


Example 5

A bicycle peptide designated 55-03-05-N317 was made corresponding to the bicycle region of the peptide ligand of Reference Example 2, with replacement of the second and third cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold.


The linear peptide used to form this bicycle was as follows:





[B-Ala][Sar]10H[dD]VPC[Aib][1Nal]G[Aib]F[1Nal][Dap]P[tBuGly]N[HArg]P[dD][Dap]


Cyclisation with TBMB was performed as described in Example 1.


The measured Ki with EphA2 was 425 nM, which demonstrates that the change to alkylamino linkages in this example preserved a significant binding affinity relative to the thioether-linked derivative of Reference Example 2.


Example 6

A bicycle peptide designated 55-03-05-N319 was made corresponding to the bicycle region of the peptide ligand of Reference Example 2, with replacement of the second and third cysteine residues by DAP residues forming alkylamino linkages to the TBMB scaffold.


The linear peptide used to form this bicycle was as follows:





[B-Ala][Sar]10H[dD]VP[Dap][Aib][1Nal]G[Aib]F[1Nal]CP[tBuGly]N[HArg]P[dD][Dap]


Cyclisation with TBMB was performed as described in Example 1.


The measured Ki with EphA2 was 17 nM, which demonstrates that the change to alkylamino linkages in this example marginally increases the affinity to EphA2 relative to the thioether-linked derivative of Reference Example 2.


Reference Examples A1-A308

The following reference peptide ligands having a TBMB scaffold with three thioether linkages to cysteine residues of the specified peptide sequences were prepared and evaluated for affinity to EphA2 as described in detail in our earlier application GB201721265.5 filed 19 Dec. 2017.


In view of the results obtained above in Examples 1-6, it is predicted that derivatives of the reference examples A1-A308 according to the present invention, i.e. having alkylamino linkages in place of one or more of the thioether linkages in the reference examples, will also display affinity for EphA2. It is further predicted that derivatives of the reference examples Bl-B98 having scaffolds other than TBMB, in particular aromatic scaffolds other than TBMB, will also display affinity for EphA2. All such derivatives having affinity for EphA2 are therefore included within the scope of the present invention.









TABLE 3







Biological Assay Data for Reference Peptide Ligands (Direct Binding Assay)










Bicycle





Compound


KD, nM ± 95% CI











Number
Sequence
Scaffold
Human EphA2
Mouse EphA2














1
ACMNDWLCSLGWTCA-Sar6-K(F1)
TBMB
107.58 ± 40.83
301 n = 1





2
AF488-G-Sar10-ACMNDWLCSLGWTC
TBMB
326 n = 1






3
ACMNDWLCELGWTCA-Sar6-K(F1)
TBMB
121.48 ± 50.27






4
ACTRQGIWCALGFEPCA-Sar6-K(F1)
TBMB
 163.5 ± 22.54






5
ACMNDWLCTLGWSCA-Sar6-K(F1)
TBMB
 142.5 ± 83.3






6
ACMNDWLCQLGWTCA-Sar6-K(F1)
TBMB
 54.25 ± 4.8






7
ACMNDWLCTLGWTCA-Sar6-K(F1)
TBMB
 74.35 ± 15.97






8
ACMNDWLCDLGWRCA-Sar6-K(F1)
TBMB
 118.5 ± 22.54






9
ACMNDWLCELGWSCA-Sar6-K(F1)
TBMB
 137.5 ± 49.98






10
ACRVSPEYCPFGPVWCAGAAA-Sar6-K(F1)
TBMB
135.13 ± 59.02






11
F1-G-Sar5-ACPWGPAWCPVHGKTCA
TBMB
   263 ± 213.64






12
F1-G-Sar5-ACPWGPAWCPVNRPGCA
TBMB
 27.78 ± 8.35






13
Ac-ACPWGPAWCPVNRPGCAGAAA-K(F1)
TBMB
    29 ± 2.55






14
AF488-G-Sar10-ACPWGPAWCPVNRPGCA
TBMB
 38 n = 1






15
F1-G-Sar5-ACPWGPMWCPVNRPGCA
TBMB
  12.6 ± 2.55






16
F1-G-Sar5-ACPWGPNWCPVNRPGCA
TBMB
  11.5 ± 1.76






17
F1-G-Sar5-AGEMACPWGPFWCPVNRPGCA
TBMB
  3.85 ± 0.1






18
F1-G-Sar5-ADVTCPWGPFWCPVNRPGCA
TBMB
  0.93 ± 0.23
4.02 ± 2





19
F1-G-Sar5-ADVRTCPWGPFWCPVNRPGCA
TBMB
  4.74 ± 0.51






20
F1-G-Sar5-ANDVTCPWGPFWCPVNRPGCA
TBMB
  2.35 ± 0.49






21
ACVPQGIWCALQFEPCA-Sar6-K(F1)
TBMB
  59.5 ± 12.78






22
ACQKQGLWCALGFEPCA-Sar6-K(F1)
TBMB
   289 ± 74.51






23
ACLVNDDCFYMGLCA-Sar6-K(F1)
TBMB
109.38 ± 20.75
















TABLE 4







Biological Assay Data for Reference Peptide Ligands (Competition Binding Assay)










Bicycle


Human EphA2 (Ki, nM ±+0 95% CI)


Compound


Fluorescent Peptide













Number
Sequence
Scaffold
Compound 66
Compound 1
Compound 12
Compound 18





24
ACMNDWLCSLGWTCA
TBMB
 82.34 ± 12.8








25
Ac-CANDWLCSLGWTC
TBMB
 328 n = 1








26
Ac-CMNDWLCALGWTC
TBMB
  71.6 ± 3.33








27
Ac-CMNDWLCSAGWTC
TBMB
 356 n = 1








28
ACMNDWLCQLGWKCA
TBMB
 113 n = 1








29
ACMNDWLCELGWTCA
TBMB
 134.5 ± 32.34








30
ACMNDWLCQLGWTCA
TBMB
 56.05 ± 3.23








31
ACTQNDWLCSLGWTCA
TBMB
151.65 ± 161.4








32
ACRNIPTMCPFGPVWCA
TBMB

83.4 n = 1







33
ACRVSPEYCPFGPVWCA
TBMB
 78.53 ± 35.61








34
ACRVSPEYCPFGPVWCAGAAA
TBMB
  77.4 ± 8.95








35
ACRVSPEYCPFGPTWCA
TBMB
  43.2 ± 13.33








36
ACRVSPEYCPFGPSWCA
TBMB
  40.5 ± 5.88








37
ACRVSPEYCPFGPEWCA
TBMB
 61.25 ± 41.85








38
ACRVSPEYCPFGPYWCA
TBMB
 26.53 ± 16.92








39
ACRVSPEYCPFGPLWCA
TBMB
 32.11 ± 10.28








40
ACRVSPEYCPFGPDWCA
TBMB
  55.4 ± 9.41








41
ACPWGPAWCPVHGKTCA
TBMB

 263 n = 1







42
ACPWGPAWCPVRDTNCA
TBMB
 316 n = 1








43
ACPWGPAWCPVNGARCA
TBMB
 430 n = 1








44
ACPWGPAWCPVNRPGCA
TBMB
191.22 ± 29.47
 164 n = 1
128.45 ± 28.21






45
ACPWGPAWCPVNRPGCAGAA
TBMB
117.13 ± 17.96

 99.15 ± 48.71




A










46
ACPWGPMWCPVNRPGCA
TBMB
 95.75 ± 29.89








47
ACPWGPNWCPVNRPGCA
TBMB
 78.35 ± 12.64








48
ACPWGPAWCPVRNPCA
TBMB
   284 ± 47.04








49
ACPWGPAWCPVSRVCA
TBMB
   428 ± 99.96








50
ACPWGPAWCPVRSCA
TBMB
   314 ± 248.92








51
ACPWGPAWCPVKPTCA
TBMB
 318.5 ± 255.78








52
ACPWGPAWCPVNRNGCA
TBMB
   168 ± 72.52








53
AGEMACPWGPFWCPVNRPGC

     6 ± 5.54


12.33 ± 2



A
TBMB









54
AVHIPCPWGPSWCPVNRPGCA
TBMB
  5.17 ± 2.76


 5.13 ± 1.52





55
AEGLPCPWGPFWCPVNRPGCA
TBMB
  6.15 ± 3.43


 11.3 ± 2.04





56
ADHACPWGPFWCPVNRPGCA
TBMB
  5.87 ± 5.09


14.43 ± 6.28





57
ADVHCPWGPFWCPVNRPGCA
TBMB
 1.2 n = 1


 0.48 ± 0.15





58
ADVTCPWGPFWCPVNRPGCA
TBMB
  2.65 ± 1.08


 1.35 ± 0.23





59
AHDVPCPWGPFWCPVNRPGC




 0.54 ± 0.14



A
TBMB









60
ADVRTCPWGPFWCPVNRPGC

 2.5 n = 1


12.63 ± 1.29



A
TBMB









61
ANDVTCPWGPFWCPVNRPGC

 7.3 n = 1


 2.93 ± 0.07



A
TBMB









62
ARDDPCPWGPFWCPVNRPGC

 27.96 ± 16.74


16.13 ± 0.8



A
TBMB









63
ACVPQGIWCALQFEPCA
TBMB
 82.45 ± 27.07
 144 n = 1
 92.2 ± 21.17






64
ACTTGSIWCALQFEPCA
TBMB
63.4 n = 1

410 n = 1






65
ACVPQGIWCALRYEPCA
TBMB
 293 n = 1

229 n = 1
















TABLE 5







Biological Assay Data for Reference Peptide Ligands (Direct Binding Assay)










Bicycle


KD, nM ± 95% CI











Compound


Human
Mouse


Number
Sequence
Scaffold
EphA2
EphA2





66
F1-G-Sar5-ACPWGPFWCPVNRPGCA
TBMB
 8.45 ± 0.4
  22 n = 1





67
AlexaFluor488-G-Sar5-
TBMB
15.03 ± 1.72
51.8 ± 6.27



ACPWGPFWCPVNRPGC








68
AlexaFluor488-(β-Ala)-Sar10-
TBMB
15.37 ± 2.87
23.4 n = 1



ACPWGPFWCPVNRPGC
















TABLE 6







Biological Assay Data for Reference Peptide Ligands (Competition Binding Assay)













Ki, nM ± 95% CI


Bicycle


Human EphA2


Compound


Fluorescent Peptide











Number
Sequence
Scaffold
Compound 18
Compound 66














69
ACPWGPFWCPVNRPGCA
TBMB
106.75 ± 44.25
 70.08 ± 8.01





70
Sar2-ACPWGPFWCPVNRPGC
TBMB
 51.81 ± 21.75
 20.45 ± 12.84





71
Ac-Sar2-ACPWGPFWCPVNRPGC
TBMB
 11.87 ± 7.51






72
(β-Ala)-Sar10-ACPWGPFWCPVNRPGC
TBMB
  29.1 ± 5.08
 20.98 ± 2.18





73
Sar2-AC(HyP)WGPFWCPVNRPGC
TBMB
  47.6 ± 18.42
 247.5 ± 18.62





74
Sar2-AC(Aib)WGPFWCPVNRPGC
TBMB

 138.9 ± 88.79





75
Sar2-AC(4F1Pro)WGPFWCPVNRPGC
TBMB

399.67 ± 90.63





76
Sar2-ACP(1Nal)GPFWCPVNRPGC
TBMB
   3.5 ± 1.96
  16.7 ± 9.68





77
Sar2-ACP(2Nal)GPFWCPVNRPGC
TBMB

458.33 ± 222.44





78
Sar2-ACPWG(Aze)FWCPVNRPGC
TBMB

 403.5 ± 12.74





79
Sar2-ACPWG(HyP)FWCPVNRPGC
TBMB

   131 ± 22.97





80
Sar2-ACPWG(Aib)FWCPVNRPGC
TBMB
 120.5 ± 81.34
186.73 ± 94.37





81
Sar2-ACPWG(4F1Pro)FWCPVNRPGC
TBMB

   294 ± 99.6





82
Sar2-ACPWG(Pip)FWCPVNRPGC
TBMB

497.33 ± 223.62





83
Sar2-ACPWGPAWCPVNRPGC
TBMB
199 n = 1
 287.5 ± 197.95





84
Sar2-ACPWGP(4Pal)WCPVNRPGC
TBMB
  33.5 ± 0.98
 81.47 ± 68.95





85
Sar2-ACPWGP(4BrPhe)WCPVNRPGC
TBMB

 174.5 ± 20.58





86
Sar2-ACPWGP(4MeoPhe)WCPVNRPGC
TBMB

 274.5 ± 36.26





87
Sar2-ACPWGP(HPhe)WCPVNRPGC
TBMB
162 n = 1
 281.2 ± 154.82





88
Sar2-ACPWGP(4,4-BPA)WCPVNRPGC
TBMB

182.67 ± 99.5





89
Sar2-ACPWGP(NO2Phe5)WCPVNRPGC
TBMB

 289.5 ± 93.1





90
Sar2-ACPWGP(3,4-DCPhe)WCPVNRPGC
TBMB

   361 ± 25.48





91
Sar2-ACPWGPYWCPVNRPGC
TBMB

137.63 ± 104.2





92
Sar2-ACPWGP(3Pal)WCPVNRPGC
TBMB

   165 ± 27.44





93
Sar2-ACPWGP(Phg)WCPVNRPGC
TBMB

 411.5 ± 128.38





94
Sar2-ACPWGP(1Nal)WCPVNRPGC
TBMB

 196.5 ± 6.86





95
Sar2-ACPWGP(2Nal)WCPVNRPGC
TBMB

 362.5 ± 110.74





96
Sar2-ACPWGPF(1Nal)CPVNRPGC
TBMB
  31.3 ± 24.11
 68.13 ± 35.66





97
Sar2-ACPWGPFWC(Aze)VNRPGC
TBMB

   286 ± 109.76





98
Sar2-ACPWGPFWC(HyP)VNRPGC
TBMB

163.33 ± 38.41





99
Sar2-ACPWGPFWC(4F1Pro)VNRPGC
TBMB

 269.5 ± 6.86





100
Sar2-ACPWGPFWCP(tBuGly)NRPGC
TBMB
  58.3 ± 50.37
112.45 ± 73.38





101
Sar2-ACPWGPFWCPVARPGC
TBMB
293 n = 1
   265 ± 235.04





102
Sar2-ACPWGPFWCPV(D-Ala)RPGC
TBMB
   317 ± 168.56
311.67 ± 195.55





103
Sar2-ACPWGPFWCPVN(HArg)PGC
TBMB
   126 ± 9.8
169.43 ± 94.28





104
Sar2-ACPWGPFWCPVNRAGC
TBMB
124 n = 1
193.67 ± 112.76





105
Sar2-ACPWGPFWCPVNR(D-Ala)GC
TBMB

470.67 ± 221.53





106
Sar2-ACPWGPFWCPVNR(Aze)GC
TBMB

   155 ± 47.04





107
Sar2-ACPWGPFWCPVNR(HyP)GC
TBMB
48.7 n = 1
 85.83 ± 57.98





108
Sar2-ACPWGPFWCPVNR(Pip)GC
TBMB

 374.5 ± 12.74





109
Sar2-ACPWGPFWCPVNR(4F1Pro)GC
TBMB

 184.5 ± 20.58





110
Sar2-ACPWGPFWCPVNR(Aib)GC
TBMB
    75 ± 13.72
139.53 ± 103.98





111
Sar2-ACPWGPFWCPVNRPAC
TBMB
108 n = 1
 237.5 ± 164.92





112
Sar2-ACPWGPFWCPVNRP(D-Ala)C
TBMB
    89 ± 15.68






113
Sar2-AC(Aib)(1Nal2)GPFWCPVNRPGC
TBMB
 10 n = 1
6.6 n = 1





114
Sar2-AC(Aib)WGPF(1Nal)CPVNRPGC
TBMB
 21 n = 1
 43 n = 1





115
Sar2-ACP(1Nal)GPFWCPV(D-Ala)RPGC
TBMB
  12.5 ± 0.98
  1.64 ± 2.48





116
Sar2-ACP(1Nal)GPFWCPVNRP(D-Ala)C
TBMB
2.95 ± 1.67
3.2 n = 1





117
Sar2-ACPWGPF(1Nal)CPV(D-Ala)RPGC
TBMB
 53 n = 1
 75 n = 1





118
Sar2-ACPWGPF(1Nal)CPVNRP(D-Ala)C
TBMB
 37 n = 1
    18 ± 13.72





119
Sar2-ACP(1Nal)G(Aib)FWCPVNRPGC
TBMB
 21 n = 1
8.4 n = 1





120
Sar2-ACP(1Nal)GPF(1Nal)CPVNRPGC
TBMB
   1.4 ± 0.39
0.98 n = 1





121
Sar2-ACP(1Nal)GPFWCP(tBuGly)NRPGC
TBMB
  3.65 ± 0.29
  2 n = 1





122
Sar2-ACP(1Nal)GPFWCPVN(HArg)PGC
TBMB
  9.55 ± 0.69
  8 n = 1





123
Sar2-ACPWG(Aib)F(1Nal)CPVNRPGC
TBMB
 63 n = 1
 46 n = 1





124
Sar2-AC(Aib)(1Nal)GPFWCPV(D-Ala)RPGC
TBMB
 26 n = 1
2.5 n = 1





125
Sar2-AC(Aib)(1Nal)GPFWCPVNRP(D-Ala)C
TBMB
   6.4 ± 0.78
  0.61 ± 0.96





126
Sar2-ACP(1Nal)G(Aib)FWCP(tBuGly)NRPGC
TBMB
 15 n = 1
 19 n = 1





127
Sar2-ACP(1Nal)G(Aib)FWCPV(D-Ala)RPGC
TBMB
 40 n = 1
 33 n = 1





128
Sar2-ACP(1Nal)G(Aib)FWCPVNRP(D-Ala)C
TBMB
 15 n = 1
 16 n = 1





129
Sar2-ACP(1Nal)GPFWCP(tBuGly)(D-Ala)RPGC
TBMB
 23 n = 1
 15 n = 1





130
Sar2-ACP(1Nal)GPFWCP(tBuGly)N(HArg)PGC
TBMB
  0.29 ± 0.34






131
Sar2-ACP(1Nal)GPFWCP(tBuGly)NR(Aib)GC
TBMB
 11 n = 1
6.8 n = 1





132
Sar2-ACP(1Nal)GPFWCP(tBuGly)NRP(D-Ala)C
TBMB
   7.7 ± 1.96
8.7 n = 1





133
Sar2-ACP(1Nal)GPFWCPV(D-Ala)(HArg)PGC
TBMB
 14 n = 1
3.7 n = 1





134
Sar2-ACP(1Nal)GPFWCPVN(HArg)P(D-Ala)C
TBMB
1.2 n = 1
  6.15 ± 0.29





135
Sar2-AC(Aib)(1Nal)G(Aib)FWCPVNR(Aib)GC
TBMB
 43 n = 1
 30 n = 1





136
Sar2-ACP(1Nal)G(Aib)FWCP(tBuGly)N(HArg)PGC
TBMB
 23 n = 1
 15 n = 1





137
Sar2-ACP(1Nal)G(Aib)FWCP(tBuGly)NR(Aib)GC
TBMB
 20 n = 1
 18 n = 1





138
Sar2-ACP(1Nal)GPFWCP(tBuGly)N(HArg)(Aib)GC
TBMB
5.1 n = 1
















TABLE 7







Biological Assay Data for Reference Peptide Ligands (Direct Binding Assay)










Bicycle


KD, nM ± 95% CI











Compound


Human
Mouse


Number
Sequence
Scaffold
EphA2
EphA2





139
AF488-(β-Ala)-Sar10-H(D-Asp)VPCPWGPFWCPVNRPGCA
TBMB
0.31 ± 0.18
 0.8 ± 0.54





140
AF488-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
2.05 ± 0.62
4.55 ± 1.04



C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








141
AF488-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
2.65 ± 0.64
 6.5 ± 0.63



C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








142
F1-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
 1.7 n = 1




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








143
AF488-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
 1.4 ± 1.46
4.69 ± 4.15



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








144
AF488-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
1.04 n = 1
2.56 n = 1



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








145
AF488-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
2.17 ± 2.08
 3.8 ± 0.55



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








146
AF488-(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
2.19 n = 1




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








147
AF488-(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
1.07 ± 0.9
3.44 ± 1.31



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C
















TABLE 8







Biological Assay Data for Reference Peptide Ligands (Competition Binding Assay)













Ki, nM ± 95% CI











Bicycle


Human EphA2
Mouse EphA2










Compound


Fluorescent peptide











Number
Sequence
Scaffold
Compound 18
Compound 18





148
Ac-Sar2-ADVH-CPWGPFWCPVNRPGC
TBMB
   1.2 ± 0.2






149
ADVH-CP(3,3-DPA)GPFWCPVNRPGCA
TBMB
  52.8 ± 11.6






150
ADVH-CP(1Nal)GPFWCPVNRPGCA
TBMB
  0.12 ± 0.07






151
ADVH-CPWAPFWCPVNRPGCA
TBMB
 393.5 ± 206.78






152
ADVH-CPWGAFWCPVNRPGCA
TBMB
   1.8 ± 0.74






153
ADVH-CPWG(Aib)FWCPVNRPGCA
TBMB
  0.51 ± 0.29






154
ADVH-CPWGPFWCAPVNRPGCA
TBMB
101.03 ± 33.68






155
ADVH-CPWGPFWCPV(D-Ala)RPGCA
TBMB
     2 ± 0.74






156
ADVH-CPWGPFWCPVN(D-Ala)PGCA
TBMB
  14.93 ± 2.3






157
Ac-Sar2-ADVT-CPWGPFWCPVNRPGC
TBMB
  0.91 ± 0.19






158
Ac-Sar2-A(D-Asp)VT-CPWGPFWCPVNRPGC
TBMB
  2.05 ± 0.42
 2.2 ± 0.4





159
Ac-Sar2-A(D-Asp)(D-Asp)T-CPWGPFWCPVNRPGC
TBMB
  2.85 ± 0.49






160
Ac-Sar2-A(D-Asp)(Cba)T-CPWGPFWCPVNRPGC
TBMB
   2.6 ± 0.11






161
Ac-Sar2-A(D-Asp)(Cpa)T-CPWGPFWCPVNRPGC
TBMB
  4.44 ± 1.08






162
Ac-Sar2-A(D-Asp)(Cpg)T-CPWGPFWCPVNRPGC
TBMB
  2.55 ± 0.55






163
Ac-Sar2-A(D-Asp)(C5g)VT-CPWGPFWCPVNRPGC
TBMB
  1.33 ± 0.27
1.74 ± 1.23





164
Ac-Sar2-AD(tBuGly)T-CPWGPFWCPVNRPGC
TBMB
  2.25 ± 0.69






165
Ac-Sar2-A(D-Asp)VT-C(AC3C)WGPFWCPVNRPGC
TBMB
   185 ± 147






166
Ac-Sar2-A(D-Asp)VT-C(AC4C)WGPFWCPVNRPGC
TBMB
  76.7 ± 73.11






167
Ac-Sar2-A(D-Asp)VT-C(AC5C)WGPFWCPVNRPGC
TBMB
138 n = 1






168
Ac-Sar2-A(D-Asp)VT-C(4BenzyPro)WGPFWCPVNRPGC
TBMB
  5.03 ± 2.24






169
Ac-Sar2-A(D-Asp)VT-C(4PhenyPro)WGPFWCPVNRPGC
TBMB
  14.4 ± 7.64






170
Ac-Sar2-A(D-Asp)VT-CP(1Nal)GPFWCPVNRPGC
TBMB
   0.6 ± 0.19






171
Ac-Sar2-A(D-Asp)VT-CPWGP(HArg)WCPVNRPGC
TBMB
  4.88 ± 2.19






172
Ac-Sar2-A(D-Asp)VT-CPWGPNWCPVNRPGC
TBMB
  3.96 ± 0.72






173
Ac-Sar2-A(D-Asp)VT-CPWGPAWCPVNRPGC
TBMB
  6.69 ± 3.49






174
Ac-Sar2-A(D-Asp)VT-CPWGPFWCPLNRPGC
TBMB
   9.1 ± 1.73






175
Ac-Sar2-A(D-Asp)VT-CPWGPFWCPVNRP(D-Asp)C
TBMB
  1.78 ± 0.54






176
Ac-Sar2-A(D-Asp)VT-CPWGPFWCPVN(HArg)P(D-Asp)C
TBMB
  4.89 ± 0.97






177
Ac-Sar2-A(D-Asp)VT-CPWGPFWCPVNR(Aib)(D-Asp)C
TBMB
  4.43 ± 2.37






178
Ac-Sar2-A(D-Asp)VT-CP(1Nal)G(Aib)FWCPVNR(Aib)GC
TBMB
   2.4 ± 0.23






179
Ac-Sar2-A(D-Asp)VT-CPWG(Aib)F(1Nal)CPVNR(Aib)GC
TBMB
  2.94 ± 0.09






180
Ac-Sar2-A(D-Asp)VT-CPWG(Aib)FWCP(tBuGly)NR(Aib)GC
TBMB
  3.83 ± 0.43






181
Ac-Sar2-A(D-Asp)VT-CP(1Nal)G(Aib)F(1Nal)CPVNR(Aib)GC
TBMB
  1.37 ± 0.41






182
Ac-Sar2-A(D-Asp)VT-
TBMB
  1.16 ± 0.39




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)PGC








183
Ac-Sar2-A(D-Asp)VT-
TBMB
  1.02 ± 0.41




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








184
Ac-Sar2-A(D-Asp)VT-
TBMB
  1.19 ± 0.29




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








185
(D-Asp)VT-CPWGPFWCPVNRPGC
TBMB
  2.17 ± 0.73






186
(D-Asp)VT-CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC
TBMB
  1.34 ± 0.18






187
AHDVP-CP(1Nal)GPFWCPVNRPGCA
TBMB
  0.32 ± 0.03






188
AHDVP-CP(1Nal)GPFWCPVNRPGC
TBMB
  1.45 ± 0.1






189
AHDVP-CPWGPF(1Nal)CPVNRPGC
TBMB
   1.3 ± 0.2






190
AHDVP-CP(1Nal)GPFWCP(tBuGly)NRPGC
TBMB
   0.7 ± 0.4






191
AHDVP-CP(1Nal)G(Aib)FWCP(tBuGly)N(HArg)PGC
TBMB
   3.1 ± 0.68






192
AHDVP-CP(1Nal)G(Aib)FWCP(tBuGly)NR(Aib)GC
TBMB
  1.75 ± 0.1






193
Ac-Sar2-AHDVP-CPWGPFWCPVNRPGC
TBMB
  0.59 ± 0.2






194
Ac-Sar2-(D-Ala)HDVP-CPWGPFWCPVNRPGC
TBMB
   1.2 ± 0.39






195
Ac-Sar2-AADVP-CPWGPFWCPVNRPGC
TBMB
  1.01 ± 0.19






196
Ac-Sar2-A(D-His)DVP-CPWGPFWCPVNRPGC
TBMB
  0.95 ± 0.24






197
Sar2-A(D-His)DVP-CPWGPFWCPVNRPGC
TBMB
 1.2






198
Ac-Sar2-A(D-His)DVCPWGPFWCPVNRPGC
TBMB
    20 ± 1.96






199
Sar2-A(D-Ala)DVP-CPWGPFWCPVNRPGC
TBMB
  3.35 ± 1.47






200
Ac-Sar2-A(D-Asp)DVP-CPWGPFWCPVNRPGC
TBMB
   4.1 ± 0.2






201
Sar2-A(Thi)DVP-CPWGPFWCPVNRPGC
TBMB
   0.6 ± 0.04






202
Sar2-A(ThiAz)DVP-CPWGPFWCPVNRPGC
TBMB
   0.7 ± 0.08






203
Sar2-A(2FuAla)DVP-CPWGPFWCPVNRPGC
TBMB
  0.49 ± 0.24






204
Ac-Sar2-A(D-His)D(tBuGly)P-CPWGPFWCPVNRPGC
TBMB
  2.15 ± 0.1






205
Sar2-AHAVP-CPWGPFWCPVNRPGC
TBMB
   1.8 ± 0.2






206
Sar2-AH(D-Ala)VP-CPWGPFWCPVNRPGC
TBMB
   8.3 ± 0.78






207
Sar2-AHEVP-CPWGPFWCPVNRPGC
TBMB
   1.3 ± 0.39






208
Sar2-AH(D-G1u)VP-CPWGPFWCPVNRPGC
TBMB
     2 ± 0.39






209
Sar2-AH(D-Asp)VP-CPWGPFWCPVNRPGC
TBMB
  1.25 ± 0.29






210
Ac-Sar2-AH(D-Asp)VP-CPWGPFWCPVNRPGC
TBMB
   1.1 ± 0.2






211
Ac-Sar2-AH(D-Asp)(tBuGly)P-CPWGPFWCPVNRPGC
TBMB
   3.1 ± 0.2






212
Ac-Sar2-AH(D-Asp)V(Sar)-CPWGPFWCPVNRPGC
TBMB
  4.95 ± 1.86






213
Ac-Sar2-AH(D-Asp)V(Aib)-CPWGPFWCPVNRPGC
TBMB
   1.9 ± 0.2






214
Sar2-AHDAP-CPWGPFWCPVNRPGC
TBMB
  22.5 ± 2.94






215
Sar2-AHD(D-Ala)P-CPWGPFWCPVNRPGC
TBMB
    26 ± 7.84






216
Sar2-AHD(Aib)P-CPWGPFWCPVNRPGC
TBMB
  2.77 ± 0.24






217
Sar2-AHD(tBuGly)P-CPWGPFWCPVNRPGC
TBMB
0.49 n = 1






218
Sar2-AHDVA-CPWGPFWCPVNRPGC
TBMB
  1.27 ± 0.07






219
Sar2-AHDV(D-Ala)-CPWGPFWCPVNRPGC
TBMB
    15 ± 3.92






220
Sar2-AHDV(Aib)-CPWGPFWCPVNRPGC
TBMB
  0.83 ± 0.15






221
Sar2-AHDV(Aze)-CPWGPFWCPVNRPGC
TBMB
   3.1 ± 0.39






222
Sar2-AHDV(Pip)-CPWGPFWCPVNRPGC
TBMB
   3.4 ± 0.2






223
(β-Ala)-Sar10-HDVP-CPWGPFWCPVNRPGC
TBMB
  1.29 ± 0.42






224
Ac-Sar2-(D-His)DVP-CPWGPFWCPVNRPGC
TBMB
  1.09 ± 0.13






225
Ac-Sar2-H(D-Asp)VP-CPWGPFWCPVNRPGC
TBMB
     1 ± 0.18
2.08 ± 1.27





226
(β-Ala)-Sar10-H(D-Asp)VP-CPWGPFWCPVNRPGC
TBMB
  0.84 ± 0.24






227
Ac-Sar2-AH(D-Asp)VP-CP(1Nal)GPFWCP(tBuGly)N(HArg)PGC
TBMB
  0.75 ± 0.36






228
Ac-Sar2-A(D-His)DVP-CPWGP(TAla)WCPVNRPGC
TBMB
 210.5 ± 48.02






229
Ac-Sar2-A(D-His)DVP-CPWGPFWCP(HArg)NRPGC
TBMB
   5.1 ± 1.18






230
Ac-Sar2-A(D-His)DVP-CP(1Nal)GPFWCP(tBuGly)N(HArg)PGC
TBMB
   1.8 ± 0.78






231
Ac-Sar2-A(D-His)DVP-
TBMB
  1.93 ± 0.23




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)PGC








232
Ac-Sar2-A(D-His)DVP-
TBMB
   0.9 ± 0.68




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








233
Ac-Sar2-A(D-His)DVP-CPWG(Aib)FWCP(tBuGly)NR(Aib)GC
TBMB
   4.8 ± 0.84






234
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  3.94 ± 1.72




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NRPGC








235
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  2.58 ± 0.96




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)PGC








236
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
     3 ± 0.71




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








237
Ac-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
2.4 n = 1




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








238
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  2.83 ± 0.19




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)(Aib)(D-Asp)C








239
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  4.91 ± 2.45




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








240
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
2.41 n = 1




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








241
Ac-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  2.98 ± 0.96




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








242
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  7.77 ± 3.02




C(Aib)(1Nal)G(Aib)FWCP(tBuGly)N(HArg)P(D-Asp)C








243
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  2.83 ± 0.72




C(Aib)(1Nal)GP(HArg)(1Nal)CP(tBuGly)NR(Aib)GC








244
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
   2.8 ± 0.26




C(Aib)(1Nal)GP(HArg)(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








245
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
 11.91 ± 4.3




C(Aib)WGP(HArg)(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








246
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  31.4 ± 24.3




C(Aib)WGP(HArg)WCP(tBuGly)N(HArg)P(D-Asp)C








247
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  29.7 ± 11.76




C(Aib)WGP(HArg)WCP(tBuGly)N(HArg)P(D-Asp)C








248
(β-Ala)-Sar10-H(D-Asp)VP-C(D-
TBMB
  28.4 ± 0.78




Ala)(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








249
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.38 ± 0.46




CP(1Nal)G(Aib)(HArg)(1Nal)CP(tBuGly)NR(Aib)GC








250
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.48 ± 0.7




CP(1Nal)G(Aib)(HArg)(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








251
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.93 ± 0.62




CP(1Nal)G(Aib)(HArg)(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








252
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  0.37 ± 0.18




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NRPGC








253
Ac-S ar2-H(D-Asp)VP-
TBMB
  0.85 ± 0.82




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)PGC








254
Ac-S ar2-H(D-Asp)VP-
TBMB
  0.74 ± 0.2
0.64 ± 0.28



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








255
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.53 ± 0.58




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








256
Ac-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
0.41 n = 1




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








257
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.07 ± 0.2




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)PGC








258
Ac-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
0.54 n = 1




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








259
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  0.91 ± 0.14




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)(Aib)(D-Asp)C








260
Ac-(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  0.75 ± 0.07




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








261
Ac-Sar2-H(D-Asp)VP-CP(1Nal)G(Aib)F(1Nal)CPVNR(Aib)GC
TBMB
  0.63 ± 0.43






262
Ac-Sar2-H(D-Asp)VP-
TBMB
  0.71 ± 0.17
0.72 ± 0.31



CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








263
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  0.73 ± 0.26




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








264
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  0.61 ± 0.31




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








265
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.55 ± 0.34




CP(1Nal)G(Aib)FWCP (tBuGly)N(HArg)P(D-Asp)C








266
Ac-Sar2-H(D-Asp)VP-CP(1Nal)G(Aib)FWCPVNR(Aib)GC
TBMB
   1.6 ± 0.63






267
Ac-Sar2-H(D-Asp)VP-CPW(Aza-Gly)PFWCPVNRP GC
TBMB
  0.66 ± 0.2






268
Ac-Sar2-H(D-Asp)VP-CPWG(Aib)F(1Nal)CPVNR(Aib)GC
TBMB
  1.24 ± 0.46






269
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  1.11 ± 0.08




CPWG(Aib)F(1Nal)CP(tBuGly)N(Harg)P(D-Asp)C








270
Ac-Sar2-H(D-Asp)VP-CPWG(Aib)FWCP(tBuGly)NR(Aib)GC
TBMB
  1.52 ± 1.27






271
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  3.66 ± 1.7




CPWG(Aib)FWCP(tBuGly)N(HArg)P(D-Asp)C








272
(β-Ala)-Sar10-H(D-Asp)VP-
TBMB
  3.99 ± 0.13




CPWGP(HArg)WCP(tBuGly)N(HArg)P(D-Asp)C








273
(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
1.5 n = 1




C(Aib)(1Nal)G(Aib)F (1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








274
Ac-(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
  2.28 ± 0.69




C(Aib)(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)(D-Asp)C








275
(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
  15.9 ± 0.2




C(Aib)WG(Aib)FWCP(tBuGly)NR(Aib)(D-Asp)C








276
(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
  0.62 ± 0.27




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








277
Ac-(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
  0.53 ± 0.15




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








278
(β-Ala)-Sar10-H(D-Asp)(C5g)P-
TBMB
  0.46 ± 0.22




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








279
(β-Ala)-Sar10-H(D-Asp)(C5g)T-
TBMB
  0.59 ± 0.28




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC








280
(β-Ala)-Sar10-H(D-Asp)(C5g)T-
TBMB
  0.64 ± 0.48




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C








281
Ac-Sar2-A(D-Asp)DVT-CPWGPFWCPVNRPGC
TBMB
  5.78 ± 1.1






282
Ac-Sar2-A(D-Asp)DVT-CP(1Nal)GPFWCPVNRPGC
TBMB
  0.87 ± 0.14






283
Ac-Sar2-A(D-Asp)DVT-CP(1Nal)GPF(1Nal)CPVNRPGC
TBMB
  0.28 ± 0.08






284
Ac-Sar2-A(D-Asp)DVT-
TBMB
   3.8 ± 0.77




CP(1Nal)G(Aib)FWCP(tBuGly)N(HArg)PGC








285
Ac-Sar2-H(D-Asp)VT-
TBMB
  1.21 ± 0.29




CP(1Nal)G(Aib)F(1Nal)CP(tBuGly)NR(Aib)GC
















TABLE 9







Biological Assay Data for Reference Peptide Ligands (Ala scan peptides, Competition Binding Assay)













Ki, nM ± 95% CI


Bicycle


Human EphA2


Compound


Fluorescent Peptide











Number
Sequence
Scaffold
Compound 66
Compound 18














25
Ac-CANDWLCSLGWTC
TBMB
328 n = 1






26
Ac-CMNDWLCALGWTC
TBMB
  71.6 ± 3.33






27
Ac-CMNDWLCSAGWTC
TBMB
356 n = 1






286
Sar2-ACAWGPFWCPVNRPGC
TBMB
   886 ± 474.47






287
Sar2-ACPAGPFWCPVNRPGC
TBMB
>11000






288
Sar2-ACPWAPFWCPVNRPGC
TBMB
>28000
>1000





289
Sar2-ACPWGAFWCPVNRPGC
TBMB
  1102 ± 186.2
>1000





83
Sar2-ACPWGPAWCPVNRPGC
TBMB
 287.5 ± 197.95
199 n = 1





290
Sar2-ACPWGPFACPVNRPGC
TBMB
>7000






291
Sar2-ACPWGPFWCAVNRPGC
TBMB
>6000
>1000





292
Sar2-ACPWGPFWCPANRPGC
TBMB
 953.5 ± 59.78






101
Sar2-ACPWGPFWCPVARPGC
TBMB
   265 ± 235.04
293 n = 1





293
Sar2-ACPWGPFWCPVNAPGC
TBMB
   711 ± 581.64






104
Sar2-ACPWGPFWCPVNRAGC
TBMB
193.67 ± 112.76
124 n = 1





111
Sar2-ACPWGPFWCPVNRPAC
TBMB
 237.5 ± 164.92
108 n = 1





294
Sar2-AC(D-Ala)WGPFWCPVNRPGC
TBMB
>4000






295
Sar2-ACP(D-Ala)GPFWCPVNRPGC
TBMB
>7000






296
Sar2-ACPW(D-Ala)PFWCPVNRPGC
TBMB
1003 n = 1






297
Sar2-ACPWG(D-Ala)FWCPVNRPGC
TBMB
1497 n = 1






298
Sar2-ACPWGP(D-Ala)WCPVNRPGC
TBMB
>6500






299
Sar2-ACPWGPF(D-Ala)CPVNRPGC
TBMB
>4000






300
Sar2-ACPWGPFWC(D-Ala)VNRPGC
TBMB
>1200






301
Sar2-ACPWGPFWCP(D-Ala)NRPGC
TBMB
>4000






102
Sar2-ACPWGPFWCPV(D-Ala)RPGC
TBMB
311.67 ± 195.55
   317 ± 168.56





302
Sar2-ACPWGPFWCPVN(D-Ala)PGC
TBMB
  1410 ± 680.11
>1000





105
Sar2-ACPWGPFWCPVNR(D-Ala)GC
TBMB
470.67 ± 221.53
677 n = 1





112
Sar2-ACPWGPFWCPVNRP(D-Ala)C
TBMB
109.83 ± 66.19
    89 ± 15.68





303
(β-Ala)-Sar10-ACPWAPFWCAVNRPGC


>1000





304
4-(pyridyl-2-disulfanyl)-4-RS -methylbutanoyl-
TBMB

>10000



(β-Ala)-Sar10-ACPWAPFWCAVNRPGC








173
Ac-Sar2-A(D-Asp)VTCPWGPAWCPVNRPGC
TBMB

  6.69 ± 3.49





305
(β-Ala)-Sar10-H(D-
TBMB

>5000



Asp)VP CP(1Nal)A(Aib)F(1Nal)CA(tBuGly)NR(Aib)(D-






Asp)C








151
ADVHCPW(Ala)PFWCPVNRPGCA
TBMB

 393.5 ± 206.78





152
ADVHCPWG(Ala)FWCPVNRPGCA
TBMB

   1.8 ± 0.74





154
ADVHCPWGPFWC(D-Ala)VNRPGCA
TBMB

101.03 ± 33.68





155
ADVHCPWGPFWCPV(D-Ala)RPGCA
TBMB

     2 ± 0.74





156
ADVHCPWGPFWCPVN(D-Ala)PGCA
TBMB

 14.93 ± 2.3





306
DOTA-(β-Ala)-Sar10-H(D-
TBMB

>250



Asp)VPCP(1Nal)A(Aib)F(1Nal)CA(tBuGly)NR(Aib)(D-






Asp)C
















TABLE 10







Biological Assay Data for Reference Peptide Ligands (Ala scan peptides, Direct Binding Assay)










Bicycle





Compound


KD, nM ± 95% CI


Number
Sequence
Scaffold
Human EphA2





307
AF488-(β-Ala)-Sar10-ACPWAPFWCAVNRPGC
TBMB
>1000





308
AF488-(β-Ala)-Sar10-H(D-
TBMB
>2000



Asp)VPCP(1Nal)A(Aib)F(1Nal)CA(tBuGly)NR(Aib)(D-Asp)C









Reference Examples B1-B98

The following reference peptide ligands having a TATA scaffold with three thioether linkages to cysteine residues of the specified peptide sequences were prepared and evaluated for affinity to EphA2 as described in detail in our earlier application GB201721259.8 filed 19 Dec. 2017.


In view of the results obtained above in Examples 1-6, it is predicted that derivatives of the reference examples B1-B98 according to the present invention, i.e. having alkylamino linkages in place of one or more of the thioether linkages in the reference examples, will also display affinity for EphA2. It is further predicted that derivatives of the reference examples B1-B98 having scaffolds other than TATA, in particular non-aromatic scaffolds other than TATA, will also display affinity for EphA2. All such derivatives having affinity for EphA2 are therefore included within the scope of the present invention.









TABLE 11







Biological Assay Data for Reference Peptide Ligands (TATA peptides, Direct Binding Assay)










Bicycle





Compound


Human EphA2 (KD,


Number
Sequence
Scaffold
nM ± 95% CI)













1
ACMNDWWCAMGWKCA-Sar6-K(F1)
TATA
   304 ± 91.99





2
ACVPDRRCAYMNVCA-Sar6-K(F1)
TATA
 74.91 ± 6.6





3
ACVVDGRCAYMNVCA-Sar6-K(F1)
TATA
 129.8 ± 80.75





4
ACVVDSRCAYMNVCA-Sar6-K(F1)
TATA
 124.6 ± 51.74





5
ACVPDSRCAYMNVCA-Sar6-K(F1)
TATA
 93.95 ± 23.62





6
ACYVGKECAIRNVCA-Sar6-K(F1)
TATA
 168.5 ± 20.58





7
ACYVGKECAYMNVCA-Sar6-K(F1)
TATA
149.73 ± 39.2





8
F1-G-Sar5-ACYVGKECAYMNVCA
TATA
218.33 ± 10.51





9
F1-(β-Ala)-Sar10-ARDCPLVNPLCLHPGWTC
TATA
  6.43 ± 1.15





10
F1-(β-Ala)-Sar10-A(HArg)DCPLVNPLCLHPGWTC
TATA
  9.07 ± 2.49





11
Ac-CPLVNPLCLHPGWTCLHG-Sar6-(D-K[F1])
TATA
  3.08 ± 0.43





12
Ac-CPLVNPLCLHPGWTCL(D-His)G-Sar6-(D-K[F1])
TATA
 10.56 ± 0.77





13
Ac-CPLVNPLCLHPGWSCRGQ-Sar6-(D-K[F1])
TATA
  5.29 ± 0.79





14
Ac-CPLVNPLCLHPGWSC(HArg)GQ-Sar6-(D-K[F1])
TATA
  9.96 ± 0.55
















TABLE 12







Biological Assay Data for Peptide Ligands of the Invention (TATA peptides, Competition Binding


Assay)













Ki, nM ± 95% CI















Mouse





Human EphA2
EphA2













Fluorescent Peptide













Bicycle



Reference
Reference
Reference


Compound


Reference
Compound
Compound
Compound


Number
Sequence
Scaffold
Compound C
B
A
C





15
ACMNDWWCAMGWKCA
TATA
 277.5 ± 38.22








16
ACVPDRRCAYMNVCA
TATA
 69.97 ± 8.67








17
(β-Ala)-Sar10-ACVPDRRCAYMNVC
TATA
 85.05 ± 1.08








18
DLRCGGDPRCAYMNVCA
TATA
  70.8 ± 2.35








19
SRPCVIDSRCAYMNVCA
TATA
 94.75 ± 24.01








20
ESRCSPDARCAYMNVCA
TATA
 57.05 ± 4.61








21
HSGCRPDPRCAYMNVCA
TATA
 62.15 ± 4.61








22
GSGCKPDSRCAYMNVCA
TATA
 63.25 ± 13.82








23
ETVCLPDSRCAYMNVCA
TATA
   130 ± 15.68








24
GQVCIVDARCAYMNVCA
TATA
 168.5 ± 16.66








25
ACVPDRRCAFENVCVDH
TATA
  97.3 ± 3.33








26
ACVPDRRCAFMNVCEDR
TATA
 39.05 ± 10.29








27
ACVPDRRCAFQDVCDHE
TATA
159 n = 1








28
ACVPDRRCAFRDVCLTG
TATA
1700 n = 1








29
ACYVGKECAYMNVCA
TATA
 209.5 ± 110.74
106.65 ± 24.94
 87.7








n = 1






30
ACQPSNHCAFMNYCA
TATA
293 n = 1
186.53 ± 86.86
137








n = 1






31
ACSPTPACAVQNLCA
TATA
223 n = 1
   177 ± 60.76







32
ACTSCWAYPDSFCA
TATA
   232 ± 52.19

151








n = 1






33
ACTKPTGFCAYPDTICA
TATA
 268.5 ± 16.66








34
ACRGEWGYCAYPDTICA
TATA
 347.5 ± 57.82








35
ACRNWGMYCAYPDTICA
TATA
 282.5 ± 65.66








36
ACPDWGKYCAYPDTICA
TATA
   160 ± 1.96








37
ACRVYGPYCAYPDTICA
TATA
 294.5 ± 20.58








38
ACSSCWAYPDSVCA
TATA
400.33 ± 205.19








39
ACQSCWAYPDTYCA
TATA
321.33 ± 119.53








40
ACGFMGLEPCETFCA
TATA
 187.5 ± 20.58








41
ACGFMGLVPCEVHCA
TATA
   155 ± 9.8








42
ACGFMGLEPCEMVCA
TATA
 320.5 ± 14.7








43
ACGFMGLEPCVTYCA
TATA
 233.5 ± 20.58








44
ACGFMGLEPCELVCA
TATA
 126.8 ± 21.17








45
ACGFMGLVPCNVFCA
TATA
   142 ± 41.16








46
ACGFMGLEPCELFCA
TATA
  81.7 ± 7.06








47
ACGFMGLEPCELFCMPK
TATA
   185 ± 74.48








48
ACGFMGLEPCELYCA
TATA
 127.5 ± 14.7








49
ACGFMGLEPCELYCAHT
TATA
   144 ± 17.64








50
ACGFMGLEPCEMYCA
TATA
   140 ± 45.08








51
ACGFMGLVPCELYCADN
TATA
  84.4 ± 36.46








52
ACPLVNPLCLTSGWKCA
TATA
115.33 ± 11.33








53
ACPMVNPLCLHPGWICA
TATA
  15.4 ± 3.17








54
ACPLVNPLCLHPGWICA
TATA
 15.25 ± 2.84








55
ACPLVNPLCLHPGWRCA
TATA
 20.55 ± 0.88








56
ACPLVNPLCNLPGWTCA
TATA
   184 ± 115.64








57
ACPLVNPLCLVPGWSCA
TATA
  35.4 ± 10








58
ACPLVNPLCLLDGWTCA
TATA
 38.35 ± 5.39








59
ACPLVNPLCLMPGWGCA
TATA
 114.5 ± 10.78








60
ACPLVNPLCMIGNWTCA
TATA
  96.2 ± 0.59








61
ACPLVNPLCLMTGWSCA
TATA
 241.5 ± 44.1








62
ACPLVNPLCMMGGWKCA
TATA
  67.1 ± 19.21








63
ACPLVNPLCLYGSWKCA
TATA
 59.05 ± 28.32








64
ACPLVNPLCLHPGWTCA
TATA
30 n = 1








65
ARDCPLVNPLCLHPGWTCA
TATA
  6.05 ± 1.38


  39.1 ± 0.39





66
(β-Ala)-Sar10-
TATA
  4.94 ± 1.41


  57.6 ± 24.86


(BCY6099)
ARDCPLVNPLCLHPGWTC










67
(β-Ala)-Sar10-
TATA
  8.51 ± 0.17


  61.7 ± 15.48


(BCY6014)
A(HArg)DCPLVNPLCLHPGWTC










68
Ac-ARD CPLVNPLCLHPGWTCA-
TATA
  19.3 ± 4.92


 166.5 ± 30.38



Sar6-(D-K)










69
Ac-
TATA
  17.5 ± 0.98


 164.5 ± 2.94



A(HArg)DCPLVNPLCLHPGWTCA-








Sar6-(D-K)










70
RPACPLVNPLCLHPGWTCA
TATA
 10.06 ± 2.96








71
RPPCPLVNPLCLHPGWTCA
TATA
 11.11 ± 2.25








72
KHSCPLVNPLCLHPGWTCA
TATA
 11.92 ± 6.04








73
ACPLVNPLCLHPGWTCLHG
TATA
  1.98 ± 0.49


  7.27 ± 1.09





74
Ac-CPLVNPLCLHPGWTCLHG
TATA
  1.76 ± 0.54








75
(β-Ala)-Sar10-
TATA
  2.48 ± 0.27


    18 ± 1.18



ACPLVNPLCLHPGWTCLHG










76
(β-Ala)-Sar10-
TATA
 10.01 ± 1.55


 75.15 ± 14.41



ACPLVNPLCLHPGWTCL(D-His)G










77
Ac-CPLVNPLCLHPGWTCLHG-
TATA
  5.41 ± 0.86


 48.23 ± 15.72


(BCY6019)
Sar6-(D-K)










78
Ac-CPLVNPLCLHPGWTCL(D-
TATA
  15.6 ± 4.7


115.03 ± 41.16



His)G-Sar6-(D-K)










79
ACPLVNPLCLHPG(2Na1)TCLHG
TATA
   162 ± 17.64








80
RHDCPLVNPLCLLPGWTCA
TATA
  7.11 ± 0.72








81
TPRCPLVNPLCLMPGWTCA
TATA
   9.8 ± 2.61








82
ACPLVNPLCLAPGWTCA
TATA
46.2 n = 1








83
ACPLVNPLCLAPGWTCSRS
TATA
  7.05 ± 1.11








84
ACPLVNPLCLEPGWTCA
TATA
53.9 n = 1








85
ACPLVNPLCLEPGWTCAKR
TATA
 10.95 ± 1.6








86
ACPLVNPLCLHPGWSCA
TATA
 56.15 ± 11.27








87
ACPLVNPLCLHPGWSCRGQ
TATA
  2.57 ± 0.63


  18.6 ± 0.59


(BCY6026)











88
Ac-CPLVNPLCLHPGWSCRGQ
TATA
  1.64 ± 0.75








89
(β-Ala)-Sar10-
TATA
  2.86 ± 1.29


 29.55 ± 4.61



ACPLVNPLCLHPGWSCRGQ










90
(β-Ala)-Sar10-
TATA
  5.41 ± 0.67


 47.05 ± 11.47



ACPLVNPLCLHPGWSC(HArg)GQ










91
Ac-CPLVNPLCLHPGWSCRGQ-
TATA
  5.98 ± 1.42


 49.87 ± 14.44


(BCY6042)
Sar6-(D-K)










92
Ac-
TATA
 10.56 ± 6.56


 75.27 ± 21.72



CPLVNPLCLHPGWSC(HArg)GQ-








Sar6-(D-K)










93
ACPLVNPLCLHPG(2Nal)SCRGQ
TATA
   228 ± 103.88








94
ACPLVNPLCLTPGWTCTNT
TATA
 13.25 ± 4.05








95
ACPMVNPLCLHPGWKCA
TATA
 11.91 ± 3.73








96
ACPMVNPLCLTPGWICA
TATA
 16.07 ± 4.58








97
ACPMVNPLCLHPGWTCA
TATA
    20 ± 1.02
















TABLE 13







Biological Assay Data for Peptide Ligands of the Invention (Competition Binding Assay)










Bicycle





Compound


Human EphA2


Number
Sequence
Scaffold
Reference Compound C





98
(β-Ala)-Sar10-H(D-Asp)VT-
TATA
251.5 ± 73.5



C(Aib)1Nal)G(Aib)F(1Nal)CP(tBuGly)N(HArg)P(D-Asp)C









All publications mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described aspects and embodiments of the present invention will be apparent to those skilled in the art without departing from the scope of the present invention. Although the present invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are apparent to those skilled in the art are intended to be within the scope of the following claims.

Claims
  • 1. A peptide ligand specific for EphA2 comprising a polypeptide comprising three residues selected from cysteine, L-2,3-diaminopropionic acid (Dap), N-beta-alkyl-L-2,3-diaminopropionic acid (N-AlkDap) and N-beta-haloalkyl-L-2,3-diaminopropionic acid (N-HAlkDap), with the proviso that at least one of said three residues is selected from Dap, N-AlkDap or N-HAlkDap, the said three residues being separated by at least two loop sequences, and a molecular scaffold, the peptide being linked to the scaffold by covalent alkylamino linkages with the Dap or N-AlkDap or N-HAlkDap residues of the polypeptide and by thioether linkages with the cysteine residues of the polypeptide when the said three residues include cysteine, such that two polypeptide loops are formed on the molecular scaffold.
  • 2. The peptide ligand as defined in claim 1, wherein the peptide ligand comprises an amino acid sequence selected from: A1-X1-A2-X2-A3
  • 3. The peptide ligand as defined in any preceding claim, wherein two of A1, A2 and A3 are selected from Dap, N-AlkDap or N-HAlkDap, and the third one of A1, A2 and A3 is cysteine, preferably wherein A2 is cysteine.
  • 4. The peptide ligand as defined in any preceding claim, wherein A1, A2 and A3 are each N-AlkDap or N-HAlkDap,
  • 5. The peptide ligand as defined in any preceding claim, wherein the molecular scaffold is an aromatic molecular scaffold, for example 1,3,5-tris(methylene)benzene.
  • 6. The peptide ligand as defined in claim 5, wherein the peptide ligand comprises an amino acid sequence selected from one or more of the peptide ligand sequences 1-308 listed in one or more of Tables 3 to 10, or a pharmaceutically acceptable salt thereof, with the proviso that one or more of the cysteine residues in said peptide ligand sequences 1-308 is replaced by Dap, N-AlkDap or N-HAlkDap.
  • 7. The peptide ligand as defined in any one of claims 1 to 6, wherein the peptide ligand sequence is selected from: [B-Ala][Sar]10H[dD]VPA1PWGPFWA2PVNRPGA3 or[B-Ala][Sar]10H[dD]VPA1[Aib][1Nal]G[Aib]F [1Nal]A2P[tBuGly]N[HArg]P [dD]A3
  • 8. The peptide ligand as defined in claim 7, wherein A2 is cysteine and Al and A3 are Dap, N-AlkDap or N-HAlkDap, preferably Dap.
  • 9. The peptide ligand as defined in any one of claims 1 to 4, wherein the molecular scaffold is a non-aromatic molecular scaffold, preferably selected from 1,1′,1″-(1,3,5-triazinane-1,3,5-triyl)triprop-2-en-1-one (TATA).
  • 10. The peptide ligand as defined in claim 9, which is selected from any one of Compounds 1-98 of Tables 11-13 or a pharmaceutically acceptable salt thereof, with the proviso that one or more of the cysteine residues in said peptide ligand sequences 1-98 is replaced by Dap, N-AlkDap or N-HAlkDap.
  • 11. The peptide ligand as defined in any one of claims 1 to 10, wherein the EphA2 is human EphA2.
  • 12. The peptide ligand as defined in any one of claims 1 to 11, wherein the peptide ligand is selective for human EphA2, but does not cross-react with human EphA1, EphA3 or EphA4.
  • 13. A drug conjugate comprising a peptide ligand as defined in any one of claims 1 to 12, conjugated to one or more effector and/or functional groups.
  • 14. The drug conjugate as defined in claim 13, wherein said cytotoxic agent is selected from DM1 or MMAE.
  • 15. A pharmaceutical composition which comprises the peptide ligand of any one of claims 1 to 12 or the drug conjugate of any one of claim 13 or 14, in combination with one or more pharmaceutically acceptable excipients.
  • 16. The peptide ligand as defined in any one of claims 1 to 12 or the drug conjugate as defined in any one of claim 13 or 14, for use in preventing, suppressing or treating a disease or disorder characterised by overexpression of EphA2 in diseased tissue.
  • 17. The peptide ligand as defined in any one of claims 1 to 12 or the drug conjugate as defined in any one of claim 13 or 14, for use in preventing, suppressing or treating cancer.
  • 18. The peptide ligand as defined in any one of claims 1 to 12 or the drug conjugate as defined in any one of claim 13 or 14, for use in preventing, suppressing or treating lung cancer.
  • 19. The peptide ligand as defined in any one of claims 1 to 12 or the drug conjugate as defined in any one of claim 13 or 14, for use in preventing, suppressing or treating non-small cell lung carcinoma.
Priority Claims (1)
Number Date Country Kind
1810316.8 Jun 2018 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2019/065993 6/18/2019 WO 00