Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PEPTIDES CAPABLE OF INHIBITING PROTEIN-PROTEIN INTERACTIONS AT GABAB1A SUBUNIT AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2024/115492
Kind Code:
A1
Abstract:
The present invention relates to peptides capable of inhibiting protein-protein interactions of GABABR1a receptors and uses thereof.

Inventors:
SEREIKAITE VITA (DK)
CHEN ZIYANG (DK)
REM PASCAL DOMINIC (CH)
BETTLER BERNHARD (CH)
STRØMGAARD KRISTIAN (DK)
Application Number:
PCT/EP2023/083377
Publication Date:
June 06, 2024
Filing Date:
November 28, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV COPENHAGEN (DK)
UNIV BASEL (CH)
International Classes:
C07K7/08; A61K38/00; A61K38/10; A61P25/08; A61P25/18; A61P25/30; C07K14/705
Domestic Patent References:
WO2017197253A22017-11-16
WO2018015296A12018-01-25
Other References:
TIAO JIM Y. ET AL: "The Sushi Domains of Secreted GABAB1 Isoforms Selectively Impair GABAB Heteroreceptor Function", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 283, no. 45, 7 November 2008 (2008-11-07), US, pages 31005 - 31011, XP093045311, ISSN: 0021-9258, DOI: 10.1074/jbc.M804464200
FENG MEI ET AL: "Molecular mechanism of secreted amyloid-[beta] precursor protein in binding and modulating GABA B R1a", CHEMICAL SCIENCE, vol. 12, no. 17, 5 May 2021 (2021-05-05), United Kingdom, pages 6107 - 6116, XP093045313, ISSN: 2041-6520, DOI: 10.1039/D0SC06946A
FUTAKI SHIROH ET AL: "Arginine-rich peptides: An abundant source of membrane-permeable peptides having potential as carriers for intracellular protein delivery", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 276, no. 8, 17 November 2000 (2000-11-17), pages 5836 - 5840, XP002210171, ISSN: 0021-9258, DOI: 10.1074/JBC.M007540200
J. BIOL. CHEM., vol. 243, 1969, pages 3552 - 59
PURE & APPL. CHEM., vol. 56, no. 5, 1984, pages 595 - 624
"Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY
BOWERY, N. G.BETTLER, B.FROESTL, W.GALLAGHER, J. P.MARSHALL, F.RAITERI, M.BONNER, T. I.ENNA, S. J.: "Mammalian γ-aminobutyric acids receptors: structure and function", PHARMACOL. REV., vol. 54, no. 2, 2002, pages 247 - 264, XP002289606, DOI: 10.1124/pr.54.2.247
"Pharmaceutical Formulation: Development of Peptides and Proteins", 1999, TAYLOR AND FRANCIS
HEANEY, C. F.KINNEY, J. W.: "Role of GABA(B) receptors in learning and memory and neurological disorders", NEUROSCI BIOBEHAV REV, vol. 63, 2016, pages 1 - 28
WEDIN, G. P.HORNFELDT, C. S.YLITALO, L. M.: "The clinical development of γ-hydroxybutyrate (GHB", CURR. DRUG SAF., vol. 1, no. 1, 2006, pages 99 - 106
BAY, T.EGHORN, L. F.KLEIN, A. B.WELLENDORPH, P.: "GHB receptor targets in the CNS: Focus on high-affinity binding sites", BIOCHEM PHARMACOL, vol. 87, no. 2, 2014, pages 220 - 228, XP028814347, DOI: 10.1016/j.bcp.2013.10.028
GASSMANN, M.BETTLER, B.: "Regulation of neuronal GABA receptor functions by subunit composition", NAT. REV. NEUROSCI., vol. 13, no. 6, 2012, pages 380 - 394
FRITZIUS, T.BETTLER, B.: "The organizing principle of GABA receptor complexes: Physiological and pharmacological implications", BASIC CLIN. PHARMACOL. TOXICOL., 2019
BETTLER, B.TIAO, J. Y.: "Molecular diversity, trafficking and subcellular localization of GABA receptors", PHARMACOL. THER., vol. 110, no. 3, 2006, pages 533 - 543, XP025038503, DOI: 10.1016/j.pharmthera.2006.03.006
BETTLER, B.KAUPMANN, K.MOSBACHER, J.GASSMANN, M.: "Molecular structure and physiological functions of GABA receptors", PHYSIOL. REV., vol. 84, 2004, pages 835 - 867
VIGOT, R.BARBIERI, S.BRAUNER-OSBORNE, H.TURECEK, R.SHIGEMOTO, R.ZHANG, Y. P.LUJAN, R.JACOBSON, L. H.BIERMANN, B.FRITSCHY, J. M.: "Differential compartmentalization and distinct functions of GABA(B) receptor variants", NEURON, vol. 50, no. 4, 2006, pages 589 - 601
BLEIN, S.GINHAM, R.UHRIN, D.SMITH, B. O.SOARES, D. C.VELTEL, S.MCLLHINNEY, R. A. J.WHITE, J. H.BARLOW, P. N.: "Structural analysis of the complement control protein (CCP) modules of GABA receptor 1a", J. BIOL. CHEM., vol. 279, no. 46, 2004, pages 48292 - 48306
ULRICH, D.BETTLER, B.: "GABA receptors: synaptic functions and mechanisms of diversity", CURR. OPIN. NEUROBIOL., vol. 17, no. 3, 2007, pages 298 - 303, XP022110143, DOI: 10.1016/j.conb.2007.04.001
PEREZ-GARCI, E.GASSMANN, M.BETTLER, B.LARKUM, M. E.: "The GABA isoform mediates long-lasting inhibition of dendritic Ca2+ spikes in layer 5 somatosensory pyramidal neurons", NEURON, vol. 50, no. 4, 2006, pages 603 - 616
SCHWENK, J.PEREZ-GARCI, E.SCHNEIDER, A.KOLLEWE, A.GAUTHIER-KEMPER, A.FRITZIUS, T.RAVEH, A.DINAMARCA, M. C.HANUSCHKIN, A.BILDL, W.: "Modular composition and dynamics of native GABA receptors identified by high-resolution proteomics", NAT. NEUROSCI., vol. 19, no. 2, 2016, pages 233 - 242, XP055415483, DOI: 10.1038/nn.4198
DINAMARCA, M. C.RAVEH, A.SCHNEIDER, A.FRITZIUS, T.FRUH, S.REM, P. D.STAWARSKI, M.LALANNE, T.TURECEK, R.CHOO, M.: "Complex formation of APP with GABA receptors links axonal trafficking to amyloidogenic processing", NAT. COMMUN., vol. 10, 2019, pages 1331
RICE, H. C.DE MALMAZET, D.SCHREURS, A.FRERE, S.VAN MOLLE, I.VOLKOV, A. N.CREEMERS, E.VERTKIN, I.NYS, J.RANAIVOSON, F. M.: "Secreted amyloid-β precursor protein functions as a GABA R 1 a ligand to modulate synaptic transmission", SCIENCE, vol. 363, no. 6423, 2019, pages eaao4827
CONKLIN, B. R.FARFEL, Z.LUSTIG, K. D.JULIUS, D.BOURNE, H. R.: "Substitution of three amino acids switches receptor specificity of Gq alpha to that of Gi alpha", NATURE, vol. 363, no. 6426, 1993, pages 274 - 6, XP002199064, DOI: 10.1038/363274a0
YOO, Y.; JUNG, J.; LEE, Y. N.; LEE, Y.; CHO, H.; NA, E.; HONG, J.; KIM, E.; LEE, J. S.; LEE, J. S.; HONG, C.; PARK, S. Y.; WIE, J.: "GABBR2 mutations determine phenotype in rett syndrome and epileptic encephalopathy", ANNALS OF NEUROLOGY, vol. 82, no. 3, 2017, pages 466 - 478, XP071642111, DOI: 10.1002/ana.25032
GARBUTT JC: "Approval of Baclofen for Alcohol Use Disorders in France: A Perspective From the United States", ALCOHOL AND ALCOHOLISM, vol. 55, no. 1, 2020, pages 48
Attorney, Agent or Firm:
HØIBERG P/S (DK)
Download PDF:
Claims:
Claims

1. A peptide comprising or consisting of the sequence

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

EX1 X2X3X4 XsXeW X7 X8 X9 X10 D X11 D (SEQ ID NO: 64) wherein

Xi is Aspartic acid (D) or Threonine (T),

X2 is Aspartic acid (D) or Glutamic acid (E),

Xa is S, Aspartic acid (D), Tyrosine (Y) or Phenylalanine (F),

X4 is absent, Tryptophan (W), 3-(2-naphthyl)-L-alanine (2Nala), O-ethyl-L- tyrosine (Oetyr), O-benzyl-L-tyrosine (Obtyr), or L-Cysteic acid (Cya), X5 is Isoleucine (I) or Threonine (T),

Xe is Aspartic acid (D), Tryptophan (W) or Alanine (A),

X7 is Tyrosine (Y) or Glycine (G),

Xs is Glycine (G), Proline (P) or Aspartic acid (D),

X9 is Alanine (A), Threonine (T) or Serine (S),

Xw is absent, Glycine (G), Serine (S) or Alanine (A),

X11 is Aspartic acid (D), Threonine (T) or Glutamic acid (E), or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions

2. The peptide according to claim 1 , wherein the functional variant comprises 1 or

2 individual amino acid substitutions, such as 2 individual amino acid substitutions, such as 1 individual amino acid substitution.

3. The peptide according to anyone of the preceding claims, wherein said peptide is an inhibitor of GB1a/2Rs (heterodimeric gamma-amino butyric acid (GABA) Bia I GABAB2 receptor).

4. The peptide according to anyone of the preceding claims, wherein said peptide is an inhibitor of protein-protein interactions at GB1a/2Rs.

5. The peptide according to anyone of the preceding claims, wherein Xi is D.

6. The peptide according to anyone of the preceding claims, wherein X2 is D.

7. The peptide according to anyone of the preceding claims, wherein X3 is S, D or Y, such as wherein X3 is D or Y, such as wherein X3 is D.

8. The peptide according to anyone of the preceding claims, wherein X4 is absent, W, 2-Nal, O-Ey-Y or Obtyr.

9. The peptide according to anyone of the preceding claims, wherein X4 is absent.

10. The peptide according to anyone of the preceding claims, wherein X4 is W, 2- Nal, O-Ey-Y or Obtyr, such as wherein X4 is W or 2-Nal.

11. The peptide according to anyone of the preceding claims, wherein X5 is I or T.

12. The peptide according to anyone of the preceding claims, wherein Xe is D.

13. The peptide according to anyone of the preceding claims, wherein Xs is P or D.

14. The peptide according to anyone of the preceding claims, wherein X9 is A or S, and wherein Xw is absent.

15. The peptide according to anyone of the preceding claims, wherein Xg is T and X10 is G, S.

16. The peptide according to anyone of the preceding claims, wherein Xw is G, S or absent, such as wherein Xw is G or S, such as wherein Xw is absent.

17. The peptide according to anyone of the preceding claims, wherein Xw is absent and wherein X11 is D or T, such as wherein Xw is absent and wherein X11 is D.

18. The peptide according to anyone of the preceding claims, wherein X11 is D or E and wherein Xw is G, S.

19. The peptide according to anyone of the preceding claims, wherein the peptide or functional variant thereof comprises at least 8 amino acid residues, such as 9 amino acid residues, such as 10 amino acid residues, such as 11 amino acid residues, such as 12 amino acid residues, such as 13 amino acid residues, such as 14 amino acid residues, such as 15 amino acid residues, such as 16 amino acid residues, such as 17 amino acid residues, such as 18 amino acid residues, such as 19 amino acid residues, such as 20 amino acid residues, such as 21 amino acid residues, such as 22 amino acid residues, such as 23 amino acid residues, such as 24 amino acid residues, such as 25 amino acid residues, such as 26 amino acid residues, such as 27 amino acid residues.

20. The peptide according to anyone of the preceding claims, wherein the peptide or functional variant thereof comprises between 8 and 35 amino acid residues, such as between 8 and 30 amino acid residues, such as between 8 and 27 amino acid residues, such as between 8 and 23 amino acid residues.

21. The peptide according to anyone of the preceding claims, wherein the peptide is selected from the group consisting of:

SEQ ID NO: 15: TETEFIAWGPTGDED

SEQ ID NO: 16: EDDDWIDWYDSDDD

SEQ ID NO: 17: EDDDWIDWGPTSDDD

SEQ ID NO: 18: EDDYIDWYDSDDD

SEQ ID NO: 19: EDDYIDWGPTSDDD

SEQ ID NO: 20: EDDD(2Nala)IDWYDSDDD

SEQ ID NO: 21 : EDDD(2Nala)IDWGPTSDDD

SEQ ID NO: 22: EDDYTDWYDSDDD

SEQ ID NO: 23: EDDYTDWGPTSDDD

SEQ ID NO: 24: ETEFIAWGPTADED

SEQ ID NO: 25: ETEYIAWGPTGDED

SEQ ID NO: 26: ETEFIDWGPTGDED

SEQ ID NO: 27: EDDSWIWWGPTGDED

SEQ ID NO: 28: EDDSWIWWYGADTD

SEQ ID NO: 29: EDDSWIDWGPTGDED

SEQ ID NO: 30: EDDSWIDWYGADTD

SEQ ID NO: 31 : ETEFI(Cya)WGPTGDED SEQ ID NO: 32: EDDD(Oetyr)IDWYDSDDD

SEQ ID NO: 33: EDDD(Obtyr)IDWYDSDDD

SEQ ID NO: 34: EDDD(Oetyr)IDWGPTSDDD

SEQ ID NO: 35: EDDD(Obtyr)IDWGPTSDDD or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

22. The peptide according to anyone of the preceding claims, wherein the peptide is N-terminally acetylated (Ac-N-terminus).

23. The peptide according to anyone of the preceding claims, wherein the peptide is C-terminally amidated (-NH2) or wherein the C-terminus is a carboxylic acid.

24. The peptide according to anyone of the preceding claims, wherein the peptide is conjugated to a cell-penetrating peptides (OPP).

25. The peptide according to anyone of the preceding claims, wherein the CPP is conjugated to the N-terminus or to the C-terminus of the peptide.

26. The peptide according to anyone of the preceding claims, wherein the CPP is conjugated to the peptide via a linker, such as via a peptidic linker.

27. The peptide according to anyone of the preceding claims, wherein the CPP is Tat, YGRKKRRQRRR (SEQ ID NO: 55),

R8, RRRRRRRR (SEQ ID NO: 56), R7, RRRRRRR (SEQ ID NO: 57),

R6, RRRRRR (SEQ ID NO: 58),

R5, RRRRR (SEQ ID NO: 59),

R4, RRRR (SEQ ID NO: 60),

R3, RRR, or

R2, RR.

28. The peptide according to anyone of the preceding claims, wherein the CPP is

R8, RRRRRRRR (SEQ ID NO: 56) or

R4, RRRR (SEQ ID NO: 60).

29. The peptide according to anyone of the preceding claims, wherein the peptide is selected from the group consisting of:

SEQ ID NO: 15: Ac-TETEFIAWGPTGDED-NH2

SEQ ID NO: 16: Ac-EDDDWIDWYDSDDD-NH2

SEQ ID NO: 17: Ac-EDDDWIDWGPTSDDD-NH2

SEQ ID NO: 18: Ac-EDDYIDWYDSDDD-NH2

SEQ ID NO: 19: Ac-EDDYIDWGPTSDDD-NH2

SEQ ID NO: 20: Ac-EDDD(2Nala)IDWYDSDDD-NH2

SEQ ID NO: 21: Ac-EDDD(2Nala)IDWGPTSDDD-NH2

SEQ ID NO: 22: Ac-EDDYTDWYDSDDD-NH2

SEQ ID NO: 23: Ac-EDDYTDWGPTSDDD-NH2

SEQ ID NO: 24: Ac-ETEFIAWGPTADED-NH2

SEQ ID NO: 25: Ac-ETEYIAWGPTGDED-NH2

SEQ ID NO: 26: Ac-ETEFIDWGPTGDED-NH2

SEQ ID NO: 27: Ac-EDDSWIWWGPTGDED-NH2

SEQ ID NO: 28: Ac-EDDSWIWWYGADTD-NH2

SEQ ID NO: 29: Ac-EDDSWIDWGPTGDED-NH2

SEQ ID NO: 30: Ac-EDDSWIDWYGADTD-NH2

SEQ ID NO: 31: Ac-ETEFI(Cya)WGPTGDED-NH2

SEQ ID NO: 32: Ac-EDDD(Oetyr)IDWYDSDDD-NH2

SEQ ID NO: 33: Ac-EDDD(Obtyr)IDWYDSDDD-NH2

SEQ ID NO: 34: Ac-EDDD(Oetyr)IDWGPTSDDD-NH2

SEQ ID NO: 35: Ac-EDDD(Obtyr)IDWGPTSDDD-NH2

SEQ ID NO: 36: Ac-RRRRRRRREDDDWIDWYDSDDD-NH2

SEQ ID NO: 37: Ac-RRRREDDDWIDWYDSDDD-NH2

SEQ ID NO: 38: Ac-RRREDDDWIDWYDSDDD-NH2

SEQ ID NO: 39: Ac-RRRRRRRREDDDWIDWGPTSDDD-NH2

SEQ ID NO: 40: Ac-RRRREDDDWIDWGPTSDDD-NH2

SEQ ID NO: 41: Ac-RRREDDDWIDWGPTSDDD-NH2

SEQ ID NO: 42: Ac-RRRREDDD(2Nala)IDWYDSDDD-NH2

SEQ ID NO: 43: Ac-RRRREDDD(2Nala)IDWGPTSDDD-NH2 or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

30. The peptide according to anyone of the preceding claims, wherein the peptide is a cyclic polypeptide.

31. The peptide according to anyone of the preceding claims, wherein the peptide is “head-to-tail” cyclized.

32. The peptide according to anyone of the preceding claims, wherein the peptide is “side chain-to-side chain” cyclized.

33. The peptide according to anyone of the preceding claims, wherein the peptide is “tail-to-side chain” cyclized.

34. The peptide according to anyone of the preceding claims, wherein the peptide is “side chain-to-head” cyclized.

35. The peptide according to anyone of the preceding claims, wherein the peptide is back bone cyclized.

36. The peptide according to anyone of the preceding claims, wherein the N- terminal amino acid residue is linked to the C-terminal amino acid residue via a peptide bond.

37. The peptide according to anyone of the preceding claims, wherein the N- terminal amino acid residue is linked to the C-terminal amino acid residue via a non-peptide chemical linker.

38. The peptide according to anyone of the preceding claims, wherein the peptides further comprises: a. two cysteines linked via a non-peptide chemical linker; and/or b. a cysteine linked to the N-terminal amino acid residue of the peptide.

39. The peptide according to anyone of the preceding claims, wherein the peptide further comprises two cysteines.

40. The peptide according to anyone of the preceding claims, wherein the peptide further comprise a cysteine at the N-terminal.

41. The peptide according to anyone of the preceding claims, wherein the peptide further comprises a cysteine at the C-terminal.

42. The peptide according to anyone of the preceding claims, wherein the peptide further comprises a cysteine at the N-terminal and a cysteine at the C-terminal.

43. The peptide according to anyone of the preceding claims, wherein a linker connects two cysteines within the peptide of the present disclosure.

44. The peptide according to anyone of the preceding claims, wherein a linker connects a cysteine at the N-terminal amino acid residue and a cysteine at the C-terminal amino acid residue of the peptide.

45. The peptide according to anyone of the preceding claims, wherein the nonpeptide chemical linker is:

46. The peptide according to anyone of the preceding claims, wherein the peptide is selected from the group consisting of:

79

47. The peptide according to anyone of the preceding claims, wherein the peptide is

48. The peptide according to anyone of the preceding claims, wherein the peptide is

S o

49. The peptide according to anyone of the preceding claims, wherein the peptide is

50. The peptide according to anyone of the preceding claims, wherein the peptide is

51. The peptide according to anyone of the preceding claims, wherein the peptide is

52. The peptide according to anyone of the preceding claims, wherein the peptide is

53. The peptide according to anyone of the preceding claims, wherein the peptide is conjugated to an additional moiety.

54. The peptide according to anyone of the preceding claims, wherein the peptide has binding affinity (K) to sushi domain (SD1/2) protein of 20 nM or less.

55. The peptide according to anyone of the preceding claims, wherein the peptide has membrane permeability (CP50) of 30 pM or less, such as of 10 pM or less.

56. The peptide according to anyone of the preceding claims, wherein the peptide has plasma stability (T1/2) of 12 hours or more, such as of 16 hours or more, such as of 20 hours or more, such as of 24 hours or more, such as of 30 hours or more, such as of 36 hours or more, such as of 40 hours or more, such as of 48 hours or more.

57. The peptide according to anyone of the preceding claims, wherein the peptide is capable of inhibiting an interaction between a protein and a sushi domain (SD) of GB1a at presynaptic neurons.

58. The peptide according to anyone of the preceding claims, wherein the peptide is capable of inhibiting an interaction between: sushi domain (SD) and adherence-junction associated protein 1 (AJAP1), SD and PILRa-associated neural protein (PIANP), and/or

SD and amyloid precursor protein (APP).

59. A peptide according to anyone of the preceding claims for use as a medicament.

60. A peptide according to anyone of the preceding claims for use in the treatment of neurological and/or psychiatric disorders.

61 . A peptide for use according to anyone of the preceding claims, wherein the neurological disorder is epilepsy, spasticity, schizophrenia, dystonia, neuropathic pain, narcolepsy, spasticity, cognitive deficits such as Mild cognitive impairment (MCI), autism spectrum disorder and/or substance-use disorders .

62. A peptide for use according to anyone of the preceding claims, wherein the psychiatric disorder is depression, anxiety and/or Attention deficit hyperactivity disorder (ADHD).

63. A method for treatment of neurological and/or psychiatric disorders comprising administration of a peptide according to anyone of the preceding claims to an individual in need thereof.

64. Use of a peptide according to anyone of the preceding claims for the manufacture of a medicament for the treatment of neurological and/or psychiatric disorders.

65. A medicament for use in the treatment or prevention of neurological and/or psychiatric disorders comprising a peptide according to anyone of the preceding claims.

Description:
Peptides capable of inhibiting protein-protein interactions at GABAeia subunit and uses thereof

Technical field

The present invention relates to peptides capable of inhibiting protein-protein interactions at the GABAsia subunit of GABAB receptors and uses thereof.

Background

Synaptic transmission is a process tightly regulated by release of neurotransmitters and it is dependent on a fine-tuned balance of neuronal excitation and inhibition. Neurotransmitters are released from a presynaptic neuron and bind to receptors on a postsynaptic neuron. Synaptic transmission at the excitatory synapses is mainly mediated and regulated by ionotropic and metabotropic glutamate receptors, respectively, which are activated by the excitatory neurotransmitter glutamate. In contrast, y-amino butyric acid (GABA), the key inhibitory neurotransmitter in the central nervous system (CNS), activates ionotropic GABAA and metabotropic GABAB receptors (GBRs), which mediate and regulate inhibitory synaptic transmission, respectively. GBRs is involved in learning and memory, and their dysfunction has been implicated in neurological and psychiatric disorders such as epilepsy, spasticity, depression, anxiety, schizophrenia, Alzheimer’s disease and cognitive deficits (Heaney et al. 2016).

However, therapeutics targeting GBRs are very scarce as only two drugs, baclofen and y-hydroxybutyrate (GHB), are currently used for the treatment of spasticity (Bowery et al. 2002), alcohol use disorders (Garbutt 2020) and narcolepsy (Wedin et al. 2006; Bay et al. 2014). Unfortunately, the use of both baclofen and GHB is associated with serious adverse side effects such as sedation and muscle relaxation, which has significantly hampered their therapeutic value in the treatment of mental health disorders (Gassmann et al. 2012; Fritzius et al. 2019).

The molecular diversity of functional GBR is defined by GABAsia (GB1a), GABABW (GB1b) and GABAB2 (GB2) subunits, which can form either heterodimeric GB1a/2 or GB1b/2 receptor subtypes (Bettier et al. 2006; Bettier et al. 2004). Most importantly, GB1a and GB1b fulfil different physiological roles resulting from distinct cellular localizations (Vigot et al. 2006). GB1a and GB1b subunits are nearly identical except for a 129-residue extension at the extreme extracellular N-terminus of GB1a, referred to as the sushi domains (SDs) (Blein et al. 2004). Previous studies indicate that the major difference between the two isoforms is that they display very distinct subcellular distribution, where the GB1a/2 receptor localizes to presynaptic sites, whereas the GB1b/2 receptor localizes to postsynaptic sites (Ulrich and Bettier 2007; Perez-Garci et al. 2006). Consequently, GB1a/2 is involved in regulating vesicular neurotransmitter release (including glutamate)at presynaptic termini, while GB1b/2 activates potassium currents in the spines and dendrites.

Recent proteomic studies identified three transmembrane proteins interacting specifically with the GB1a/2 receptor: the amyloid precursor protein (APP), the adherence-junction associated protein 1 (AJAP1) and the PILRa-associated neural protein (PIANP) (Schwenk et al. 2016; Dinamarca et al. 2019; Rice et al. 2019). Sequence alignment of the APP, AJAP1 and PIANP sequences displayed no obvious similarity, except for a stretch of six amino acids, suggesting a potential central binding interface, which mediates the interaction of these proteins with the N-terminal Sushi domain 1 (SD1) of GBR1a/2. Interestingly, the affinity for SD1 binding is different within the three SD-interacting proteins and follows the order AJAP1 > PIANP » APP. Functionally, APP mediates the trafficking of GB1a/2 receptors to the presynaptic site, where GB1a/2 receptors are likely transferred to AJAP1 or PIANP that precisely localize the receptor at the cell surface. Activated presynaptic GBRs at glutamatergic terminals inhibit the release of neurotransmitters (glutamate) and thereby inhibit excitatory neuronal transmission.

Mild cognitive impairment (MCI) is a stage of cognitive decline between normal aging caused cognitive decline and dementia. Consequently, patients suffering from MCI have difficulties with memory, language, thinking or judgment. Worldwide, the annual prevalence of MCI in persons older than 60 years is estimated to be between 12-18%. Additionally, among MCI patient groups approx. 10-15% develop dementia annually, and dementia is reported as the seventh leading cause of death worldwide. Currently there is no approved treatment directed at MCI.

Cognitive functions like learning and memory depend on balanced-neuronal transmission. An increase in glutamate or acetylcholine release at excitatory synapses is expected to have cognition enhancing effects in Alzheimer’s disease and patients with MCI (Fritzius et al. 2019). Therefore, increasing excitatory neurotransmission in patients with cognition decline may offer a new mode of action to target cognitive impairments, and there is a need for new agents that can modulate excitatory neurotransmission.

Summary

The current invention is directed to compounds that inhibit GBR-associated proteinprotein interactions (PPIs), specifically those involving the N-terminal sushi domains (SDs), and in particular Sushi domain 1 (SD1), of the GABAsia/2 (GB1a/2) receptor, which is present at excitatory terminals.

The compounds disclosed herein are capable of inhibiting protein-protein interactions such as interactions between any one of adherence-junction associated protein 1 (AJAP1), PILRa-associated neural protein (PIANP) and amyloid precursor protein (APP) and the N-terminal SD1 of the GB1a/2receptor. Interactions with these proteins stabilize presynaptic GB1a/2 receptors at glutamatergic terminals, which controls inhibition of excitatory neurotransmitter release. The compounds of the present disclosure can thus upregulate excitatory neurotransmitter release, resulting in cognition enhancing effects. In particular, the compounds disclosed herein can uncouple presynaptic GB1a/2 from APP, AJAP1 and PIANP and therefore cause delocalization of the receptors and prevent inhibition of neurotransmitter release. Consequently, this will lead to an increase in excitatory neurotransmission and an enhancement of cognitive functions.

The developed peptide-based modulators act specifically at presynaptic GB1a/2 receptors, resulting in increased efficacy and specificity, and reduced adverse effects.

Thus, one aspect of the present disclosure relates to a peptide comprising or consisting of the sequence

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

E Xi X2X3X4 X5X6 W X 7 X 8 X 9 X10 D X11 D (SEQ ID NO: 64) wherein

Xi is Aspartic acid (D) or Threonine (T),

X2 is Aspartic acid (D) or Glutamic acid (E),

X 3 is S, Aspartic acid (D), Tyrosine (Y) or Phenylalanine (F),

X4 is absent, Tryptophan (W), 3-(2-naphthyl)- L-alanine (2Nala), O-ethyl-L-tyrosine (Oetyr), O-benzyl-L-tyrosine (Obtyr), or L-Cysteic acid (Cya), Xs is Isoleucine (I) or Threonine (T),

Xs is Aspartic acid (D), Tryptophan (W) or Alanine (A),

X? is Tyrosine (Y) or Glycine (G),

Xs is Glycine (G), Proline (P) or Aspartic acid (D),

X9 is Alanine (A), Threonine (T) or Serine (S),

Xw is absent, Glycine (G), Serine (S) or Alanine (A),

X11 is Aspartic acid (D), Threonine (T) or Glutamic acid (E), or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitution.

Another aspect of the present disclosure relates to a peptide defined herein for use as a medicament.

Another aspect of the present disclosure relates to a peptide defined herein for use in the treatment of neurological and/or psychiatric disorders, such as wherein the neurological disorder is epilepsy, spasticity, schizophrenia, dystonia, neuropathic pain, narcolepsy, spasticity, cognitive deficits such as Mild cognitive impairment (MCI), autism spectrum disorder and/or substance-use disorders ; and/or wherein the psychiatric disorder is depression, anxiety and/or Attention deficit hyperactivity disorder (ADHD).

Description of Drawings

Figure 1 : Ala scan of APPWT, AJAP-1WT and PIANPWT. A-C) Affinity fold-change of Ala scan variants of APPWT (A), AJAP-1WT (B) and PIANPWT (C). D) Alignment and positions numbering of APPWT, AJAP-1WT and PIANPWT. All presented peptides have an acetylated N-terminal amino acid and a C-terminal amide.

Figure 2: In silico ncAAs-based positional scanning of APPWT, AJAP-1WT and PIANPWT. A-C) The heat maps of computationally predicted relative affinity of ncAAs deep mutational scanning arrays of APPWT (A), AJAP-1WT (B) and PIANPWT (C) based on the obtained cAAs deep mutational scanning results. D) Overview of synthesized ncAAs incorporated APPWT variants including the abbreviations of introduced ncAAs and calculated Ki values. All synthesized peptides have an acetylated N-terminal amino acid and a C-terminal amide. Figure 3: Characterization of peptides in pull-down assay using the homogenized membrane fraction of wild type mouse brains. AJAP-1WT and designed high affinity peptide 20 are capable of pulling down native GB1a while the control groups including beads alone and scrambled version peptide of 20 (Scr-20) exhibit no enrichment of this protein as shown by immunoblotting.

Figure 4: Evaluation and optimization of cell membrane permeability and plasma stability of the designed peptides. A - B) Comparison of SD1/2 binding affinity (FP inhibition assay based calculated K\, mean ± SEM, n > 3) of 16 (A) and 17 (B) tagged with poly R of different lengths from 8 R to 3 R. C - D) Comparison of cell membrane permeability (CAPA assay based calculated CP50) of 16 (C) and 17 (D) tagged with poly R of different lengths from 8 R to 3 R and chloroalkane (CA) at the N-terminal. E- F) Comparison of in vitro human plasma stability (expressed as percentage of remaining peptides after 48 h endpoint incubation in human plasma) of 16 (E) and 17 (F) tagged with poly R of different lengths from 8 R to 3 R. G - H) Comparison of the effects of introduced ncAA (2Nala) on the cell membrane permeability (G) (CAPA assay based calculated CP50, mean ± SEM, n > 3) and SD1/2 binding affinity (H) (FP inhibition assay based calculated K\, mean ± SEM, n > 3) of 4 R tagged 16, 17, 20 and 21.

Figure 5: High-affinity peptides efficiently abolish the negative allosteric properties of AJAP-1 at transcellular GB1a/2 receptors. A) Assay monitoring PLC dependent FLuc (firefly luciferase) expression under control of the serum response element (SRE). GB1a/2 receptors were artificially coupled to PLC by stably expressing the chimeric G protein subunit Ga q /j. GB1a/2 and SRE-FLuc reporter were transiently expressed in HEK293T-Gagi cells. B) Peptides (AJAP-1 wr, 16 and 20) at 10 pM significantly alter dose-response curves of GABA-induced FLuc activity in transfected cells in the presence of AJAP-1 in trans (AJAP-1 expressed in neighboring cells). C) The addition of 10 pM AJAP-1WT, 16 or 20 elevate the basal activity of GB1a/2 receptors in presence of AJAP-1 in trans. D) The addition of 10 pM AJAP-1WT, 16 or 20 decreased the EC50 of GABA for activating GB1a/2 receptors in presence of AJAP-1 in trans. E) The addition of 10 pM AJAP-1 wr, 16 or 20 did not altor the maximum efficacy (Emax ) of GABA for activating GB1a/2Rs in presence of AJAP-1 in trans. Note: the vehicle is PBS.

Figure 6: High-affinity peptide efficiently abolishes AJAP1 -mediated recruitment of GB1a/2 receptors at neuron’s membrane. A) Confocal fluorescent imaging of AJAP-1 and GBRs using AJAP-1 transfected HEK293T I mouse hippocampal neuron cocultures. B) The guantification of the fluorescence intensity of GBRs at the neuronal membrane reveals a significant reduction in membrane abundance when exposed to the high-affinity peptide 20 compared to the negative control scrambled peptide Scr-20. Furthermore, the inhibitory effect becomes increasingly significant with extended incubation time at 30 minutes and 60 minutes relative to at 10 min.

Definitions

The term “affinity” refers to the strength of binding between a ligand and its receptor.

An “amino acid residue” can be a natural or non-natural amino acid residue linked by peptide bonds or bonds different from peptide bonds. The amino acid residues can be in D-configuration or L-configuration. An amino acid residue comprises an amino terminal part (NH2) and a carboxy terminal part (COOH) separated by a central part comprising a carbon atom, or a chain of carbon atoms, at least one of which comprises at least one side chain or functional group. NH2 refers to the amino group present at the amino terminal end of an amino acid or peptide, and COOH refers to the carboxy group present at the carboxy terminal end of an amino acid or peptide. The generic term amino acid comprises both natural and non-natural amino acids. Natural amino acids of standard nomenclature as listed in J. Biol. Chem., 243:3552-59 (1969) and adopted in 37 C.F.R., section 1.822(b)(2) belong to the group of amino acids listed herewith: Y,G,F,M,A,S,I,L,T,V,P,K,H,Q,E,W,R,D,N and C. Non-natural amino acids are those not listed immediately above. Also, non-natural amino acid residues include, but are not limited to, modified amino acid residues, L-amino acid residues, and stereoisomers of D-amino acid residues.

An “equivalent amino acid residue” refers to an amino acid residue capable of replacing another amino acid residue in a polypeptide without substantially altering the structure and/or functionality of the polypeptide. Equivalent amino acids thus have similar properties such as bulkiness of the side-chain, side chain polarity (polar or non-polar), hydrophobicity (hydrophobic or hydrophilic), pH (acidic, neutral or basic) and side chain organization of carbon molecules (aromatic/aliphatic). As such, “equivalent amino acid residues” can be regarded as “conservative amino acid substitutions”.

Within the meaning of the term “equivalent amino acid substitution” as applied herein, one amino acid may be substituted for another, in one embodiment, within the groups of amino acids indicated herein below: Amino acids having polar side chains (Asp, Glu, Lys, Arg, His, Asn, Gin, Ser, Thr, Pro, and Cys); Amino acids having non-polar side chains (Gly, Ala, Vai, Leu, lie, Phe, Trp, Tyr and Met); Amino acids having aliphatic side chains (Gly, Ala Vai, Leu, lie); Amino acids having cyclic side chains (Trp, His, Pro); Amino acids having aromatic side chains (Phe, Tyr, Trp); Amino acids having acidic, such as negatively charged side chains (Asp, Glu); Amino acids having basic, such as positively charged side chains (Lys, Arg, His); Amino acids having amide side chains (Asn, Gin); Amino acids having hydroxy side chains (Ser, Thr); Amino acids having sulphur-containing side chains (Cys, Met); Neutral, weakly hydrophobic amino acids (Pro, Ala, Gly, Ser, Thr); Hydrophilic, acidic amino acids (Gin, Asn, Glu, Asp); and Hydrophobic amino acids (Leu, lie, Vai).

Where the L or D form (optical isomers) has not been specified it is to be understood that the amino acid in question has the natural L form, cf. Pure & Appl. Chem. Vol. (56(5) pp 595-624 (1984) or the D form, so that the peptides formed may be constituted of amino acids of L form, D form, or a sequence of mixed L forms and D forms.

A “functional variant” of a peptide is a peptide capable of performing essentially the same functions as the peptide it is a functional variant of. In particular, a functional variant can bind the same molecules, preferably with the same affinity, as the peptide it is a functional variant of.

The terms "drug" and "medicament" as used herein include biologically, physiologically, or pharmacologically active substances that act locally or systemically in the human or animal body.

The terms “treatment” and “treating” as used herein refer to the management and care of a patient for the purpose of combating a condition, disease or disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, and refer equally to curative therapy, prophylactic or preventative therapy and ameliorating or palliative therapy, such as administration of the peptide or composition for the purpose of: alleviating or relieving symptoms or complications; delaying the progression of the condition, partially arresting the clinical manifestations, disease or disorder; curing or eliminating the condition, disease or disorder; amelioration or palliation of the condition or symptoms, and remission (whether partial or total), whether detectable or undetectable; and/or preventing or reducing the risk of acquiring the condition, disease or disorder, wherein “preventing” or “prevention” is to be understood to refer to the management and care of a patient for the purpose of hindering the development of the condition, disease or disorder, and includes the administration of the active compounds to prevent or reduce the risk of the onset of symptoms or complications. The term "palliation", and variations thereof, as used herein, means that the extent and/or undesirable manifestations of a physiological condition or symptom are lessened and/or time course of the progression is slowed or lengthened, as compared to not administering compositions of the present invention.

The term “individual” refers to vertebrates, particular members of the mammalian species, preferably primates including humans. As used herein, ‘subject’ and ‘individual’ may be used interchangeably. Treatment of animals, such as mice, rats, dogs, cats, cows, horses, sheep and pigs, is, however, also within the scope of the present invention.

An “individual in need thereof” refers to an individual who may benefit from treatment. In one embodiment, said individual in need thereof is a diseased individual, wherein said disease may be a neurological and/or a psychiatric disorder.

A "treatment effect" or "therapeutic effect" is manifested if there is a change in the condition being treated, as measured by the criteria constituting the definition of the terms "treating" and "treatment." There is a "change" in the condition being treated if there is at least 5% improvement, preferably 10% improvement, more preferably at least 25%, even more preferably at least 50%, such as at least 75%, and most preferably at least 100% improvement. The change can be based on improvements in the severity of the treated condition in an individual, or on a difference in the frequency of improved conditions in populations of individuals with and without treatment with the bioactive agent, or with the bioactive agent in combination with a pharmaceutical composition of the present invention.

A treatment according to the invention can be prophylactic, ameliorating and/or curative.

"Pharmacologically effective amount", “pharmaceutically effective amount” or "physiologically effective amount” of a “bioactive agent" is the amount of a bioactive agent present in a pharmaceutical composition as described herein that is needed to provide a desired level of active agent in the bloodstream or at the site of action in an individual (e.g. the lungs, the gastric system, the colorectal system, prostate, etc.) to be treated to give an anticipated physiological response when such composition is administered. Detailed description

Peptides

One aspect of the present disclosure relates to a peptide comprising or consisting of the sequence

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

E Xi X2X3X4 X5X6 W X 7 X 8 X 9 X10 D X11 D (SEQ ID NO: 64) wherein

Xi is Aspartic acid (D) or Threonine (T),

X2 is Aspartic acid (D) or Glutamic acid (E),

X 8 is S, Aspartic acid (D), Tyrosine (Y) or Phenylalanine (F),

X4 is absent, Tryptophan (W), 3-(2-naphthyl)- L-alanine (2Nala), O-ethyl-L-tyrosine (Oetyr), O-benzyl-L-tyrosine (Obtyr), or L-Cysteic acid (Cya), X5 is Isoleucine (I) or Threonine (T),

Xe is Aspartic acid (D), Tryptophan (W) or Alanine (A),

X 7 is Tyrosine (Y) or Glycine (G),

X 8 is Glycine (G), Proline (P) or Aspartic acid (D),

X 9 is Alanine (A), Threonine (T) or Serine (S),

Xw is absent, Glycine (G), Serine (S) or Alanine (A),

X11 is Aspartic acid (D), Threonine (T) or Glutamic acid (E), or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

One aspect of the present disclosure relates to a peptide comprising or consisting of an amino acid sequence disclosed in any one of Table 3, Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15 or Table 16, as well as functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

In one embodiment of the present disclosure, the functional variant of the peptides disclosed herein comprises 2 individual amino acid substitutions.

In one embodiment of the present disclosure, the functional variant of the peptides disclosed herein comprises 1 individual amino acid substitution. In one embodiment of the present disclosure, the peptides or functional variants thereof comprises at least 8 amino acid residues, such as at least 9 amino acid residues, such as at least 10 amino acid residues, such as at least 11 amino acid residues, such as at least 12 amino acid residues, such as at least 13 amino acid residues, such as at least 14 amino acid residues, such as at least 15 amino acid residues, such as between 8 and 35 amino acid residues, such as between 8 and 30 amino acid residues, such as between 8 and 27 amino acid residues, such as between 8 and 23 amino acid residues.

In one embodiment of the present disclosure, the peptides or functional variants thereof comprises no more than 35 amino acid residues, such as no more than 34 amino acid residues, such as no more than 33 amino acid residues, such as no more than 32 amino acid residues, such as no more than 31 amino acid residues, such as no more than 30 amino acid residues, such as no more than 29 amino acid residues, such as no more than 28 amino acid residues, such as no more than 27 amino acid residues, such as no more than 26 amino acid residues, such as no more than 25 amino acid residues, such as no more than 24 amino acid residues, such as no more than 23 amino acid residues, such as no more than 22 amino acid residues, such as no more than 21 amino acid residues, such as no more than 20 amino acid residues.

In one embodiment of the present disclosure, the peptides or functional variants thereof consists of at least 8 amino acid residues. In one embodiment of the present disclosure, the peptides or functional variants thereof consists of 8 amino acid residues, such as 9 amino acid residues, such as 10 amino acid residues, such as 11 amino acid residues, such as 12 amino acid residues, such as 13 amino acid residues, such as 14 amino acid residues, such as 15 amino acid residues, such as 16 amino acid residues, such as 17 amino acid residues, such as 18 amino acid residues, such as 19 amino acid residues, such as 20 amino acid residues, such as 21 amino acid residues, such as 22 amino acid residues, such as 23 amino acid residues, such as 24 amino acid residues, such as 25 amino acid residues, such as 26 amino acid residues, such as 27 amino acid residues.

In one embodiment of the present disclosure, Xi of the peptides disclosed herein is D. In one embodiment of the present disclosure, X20f the peptides disclosed herein is D.

In one embodiment of the present disclosure, X3 of the peptides disclosed herein is S, D or Y. In one embodiment of the present disclosure, X3 of the peptides disclosed herein is D or Y. In one embodiment of the present disclosure, X3 of the peptides disclosed herein is X3 is D.

The present inventors have found that residues at positions 5 to 8, corresponding to X4 XsXeW, of the peptides disclosed herein form most interactions with SD1 , and these interactions are facilitated by the presence of hydrophobic amino acid residues at one or more of these positions. Hence, presence of hydrophobic amino acid residues at one or more of these positions makes the peptides of the present disclosure have high affinity to SD1 and to GB1a/2Rs.

Thus, in one embodiment one or more of the amino acid residues at positions 5 to 8 of the peptides of the present disclosure, or functional variants thereof, are hydrophobic residues.

In one embodiments one or both of the amino acid residues at positions 6 and 8 of the peptides of the present disclosure, or functional variants thereof, are hydrophobic residues.

In one embodiment of the present disclosure, X4 of the peptides disclosed herein is absent, or it is W, 2-Nala, Oetyr or Obtyr. Presence of an unnatural amino acid at position 5, such as at X4 may improve affinity. Presence of an unnatural amino acid at position 5, such as at X4 may improve stability of the peptide to proteases.

In one embodiment of the present disclosure, X4 of the peptides disclosed herein is absent.

In one embodiment of the present disclosure, X40f the peptides disclosed herein is W, 2-Nala, Oetyr or Obtyr. In one embodiment of the present disclosure, X4 of the peptides disclosed herein is W or 2-Nala.

In one embodiment of the present disclosure, X5 of the peptides disclosed herein is I. In one embodiment of the present disclosure, Xs of the peptides disclosed herein is T.

In one embodiment of the present disclosure, Xs of the peptides disclosed herein is D.

The present inventors have also found that the amino acid residue at position 10, corresponding to Xs, is involved in pi-pi interactions with certain residues of SD1 , thus, presence of suitable residues at this position may also be important for the affinity of the peptides of the present disclosure to SD1 and to GB1a/2Rs.

Thus, in one embodiment of the present disclosure, Xs of the peptides disclosed herein is P or D.

In one embodiment of the present disclosure, Xg of the peptides disclosed herein is A or S, and Xw is absent.

In one embodiment of the present disclosure, Xg of the peptides disclosed herein is T and Xw is G or S.

In one embodiment of the present disclosure, Xw of the peptides disclosed herein is G, S or absent. In one embodiment of the present disclosure, Xw of the peptides disclosed herein is G or S. In one embodiment of the present disclosure, Xw is absent.

In one embodiment of the present disclosure, Xw of the peptides disclosed herein is absent and X11 is D or T. In one embodiment of the present disclosure, Xw of the peptides disclosed herein is absent and X11 is D.

In one embodiment of the present disclosure, X11 of the peptides disclosed herein is D or E and Xw is G or S.

In one embodiment of the present disclosure, the peptide is selected from the group consisting of:

SEQ ID NO: 15: TETEFIAWGPTGDED

SEQ ID NO: 16: EDDDWIDWYDSDDD

SEQ ID NO: 17: EDDDWIDWGPTSDDD SEQ ID NO: 18: EDDYIDWYDSDDD

SEQ ID NO: 19: EDDYIDWGPTSDDD

SEQ ID NO: 20: EDDD(2Nala)IDWYDSDDD

SEQ ID NO: 21 : EDDD(2Nala)IDWGPTSDDD

SEQ ID NO: 22: EDDYTDWYDSDDD

SEQ ID NO: 23: EDDYTDWGPTSDDD

SEQ ID NO: 24: ETEFIAWGPTADED

SEQ ID NO: 25: ETEYIAWGPTGDED

SEQ ID NO: 26: ETEFIDWGPTGDED

SEQ ID NO: 27: EDDSWIWWGPTGDED

SEQ ID NO: 28: EDDSWIWWYGADTD

SEQ ID NO: 29: EDDSWIDWGPTGDED

SEQ ID NO: 30: EDDSWIDWYGADTD

SEQ ID NO: 31 : ETEFI(Cya)WGPTGDED

SEQ ID NO: 32: EDDD(Oetyr)IDWYDSDDD

SEQ ID NO: 33: EDDD(Obtyr)IDWYDSDDD

SEQ ID NO: 34: EDDD(Oetyr)IDWGPTSDDD

SEQ ID NO: 35: EDDD(Obtyr)IDWGPTSDDD or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

In one embodiment of the present disclosure, the peptide is N-terminally acetylated (Ac-N-terminus).

In one embodiment of the present disclosure, the peptide is C-terminally amidated (- NH 2 ).

In one embodiment of the present disclosure, the C-terminus of any of the peptides disclosed herein is a carboxylic acid.

The trafficking process of GB1a-containing receptors along the axon localizes intracellularly, thus the cell membrane represents a significant barrier to be crossed by peptides that are intended to modulate GB1a-containing receptors and in particular to target the interaction between certain proteins and SD1/2, such as between any of APP, AJAP-1 and PIANP and SD1/2. Thus, in one embodiment of the present disclosure, the peptide disclosed herein is conjugated to a cell-penetrating peptide (CPP). Presence of a CPP may facilitate both cell penetration and permeability through the blood brain barrier.

In one embodiment of the present disclosure, the CPP may be conjugated to the N- terminus or to the C-terminus of a peptide of the present disclosure. In one embodiment of the present disclosure, the CPP may be conjugated to the N-terminus of a peptide of the present disclosure.

In one embodiment of the present disclosure, the CPP may be any one of:

Tat, YGRKKRRQRRR (SEQ ID NO: 55),

R8, RRRRRRRR (SEQ ID NO: 56),

R7, RRRRRRR (SEQ ID NO: 57),

R6, RRRRRR (SEQ ID NO: 58),

R5, RRRRR (SEQ ID NO: 59),

R4, RRRR (SEQ ID NO: 60),

R3, RRR, or

R2, RR.

Longer CPP may improve cell permeability of a peptide of the present disclosure. However, too long CPP may negatively affect affinity of a peptide of the present disclosure for SD1/2.

In one embodiment of the present disclosure, the CPP is

R8, RRRRRRRR (SEQ ID NO: 56) or

R4, RRRR (SEQ ID NO: 60).

In one embodiment of the present disclosure, the CPP may conjugated to a peptide of the present disclosure via a linker, for example via a peptidic linker.

In one embodiment of the present disclosure, the peptide is selected from the group consisting of:

SEQ ID NO: 15: Ac-TETEFIAWGPTGDED-NH 2

SEQ ID NO: 16: Ac-EDDDWIDWYDSDDD-NH 2 SEQ ID NO: 17: Ac-EDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 18: Ac-EDDYIDWYDSDDD-NH 2

SEQ ID NO: 19: Ac-EDDYIDWGPTSDDD-NH 2

SEQ ID NO: 20: Ac-EDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 21 : Ac-EDDD(2Nala)IDWGPTSDDD-NH 2

SEQ ID NO: 22: Ac-EDDYTDWYDSDDD-NH 2

SEQ ID NO: 23: Ac-EDDYTDWGPTSDDD-NH 2

SEQ ID NO: 24: Ac-ETEFIAWGPTADED-NH 2

SEQ ID NO: 25: Ac-ETEYIAWGPTGDED-NH 2

SEQ ID NO: 26: Ac-ETEFIDWGPTGDED-NH 2

SEQ ID NO: 27: Ac-EDDSWIWWGPTGDED-NH 2

SEQ ID NO: 28: Ac-EDDSWIWWYGADTD-NH 2

SEQ ID NO: 29: Ac-EDDSWIDWGPTGDED-NH 2

SEQ ID NO: 30: Ac-EDDSWIDWYGADTD-NH 2

SEQ ID NO: 31 : Ac-ETEFI(Cya)WGPTGDED-NH 2

SEQ ID NO: 32: Ac-EDDD(Oetyr)IDWYDSDDD-NH 2

SEQ ID NO: 33: Ac-EDDD(Obtyr)IDWYDSDDD-NH 2

SEQ ID NO: 34: Ac-EDDD(Oetyr)IDWGPTSDDD-NH 2

SEQ ID NO: 35: Ac-EDDD(Obtyr)IDWGPTSDDD-NH 2

SEQ ID NO: 36: Ac-RRRRRRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 37: Ac-RRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 38: Ac-RRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 39: Ac-RRRRRRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 40: Ac-RRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 41 : Ac-RRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 42: Ac-RRRREDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 43: Ac-RRRREDDD(2Nala)IDWGPTSDDD-NH 2 or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

In one embodiment of the present disclosure, the peptide is conjugated to an additional moiety. Activities of the peptides

In one embodiment, the present disclosure relates to peptides that are inhibitors of GB1a/2Rs (heterodimeric GABAsia / GABAB2 receptor).

In one embodiment, the present disclosure relates to peptides that are capable of modulating excitatory neurotransmission.

GB1a/2Rs display very distinct subcellular distribution compared to GB1b/2Rs, and in particular GB1a/2Rs are localized at presynaptic sites, whereas GB1b/2Rs accumulate at postsynaptic terminals. Consequently, the peptides disclosed herein inhibit GB1a/2Rs at presynaptic sites and in particular inhibit activation of GB1a/2Rs at presynaptic sites. Consequently, the peptides of the present disclosure may enhance excitatory neuronal transmission by preventing and/or stopping GB1a/2Rs from inhibiting the release of neurotransmitters such as glutamate.

In particular, in one embodiment, the peptides of the present disclosure, as well as functional variants thereof, are capable of inhibiting an interaction between a protein and a sushi domain 1/2 (SD1/2) of GB1a, such as an interaction between SD1 of GB1a at presynaptic neurons.

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, are capable of inhibiting an interaction between SD1 of GB1a, such as an interaction between SD1 of GB1a at presynaptic neurons.

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, are capable of inhibiting an interaction between:

SD and adherence-junction associated protein 1 (AJAP1), SD and PILRa-associated neural protein (PIANP), and/or SD and amyloid precursor protein (APP).

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, have binding affinity (Ki) to a sushi domain 1/2 (SD1/2) protein of 20 nM or less.

A definition of SD1/2 is found in Dinamarca et al. 2019. For example, the peptides of the present disclosure, as well as functional variants thereof, have higher binding affinity (K) to SD1/2 than wild-type AJAP-1 (SEQ ID NO: 15).

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, have binding affinity (K) to a sushi domain 1/2 (SD1/2) protein of 19 nM or less, such as of 18 nM or less, such as of 17 nM or less, such as of 16 nM or less, such as of 15 nM or less, such as of 14 nM or less, such as of 13 nM or less, such as of 12 nM or less, such as of 11 nM or less, such as of 10 nM or less, such as of 9 nM or less, such as of 8 nM or less, such as of 7 nM or less, such as of 6 nM or less, such as of 5 nM or less, such as of 4 nM or less, such as of 3 nM or less, such as of 2 nM or less, such as of about 1 nM.

Binding affinity can be measured according to any conventional methods for determining affinity of a peptide or ligand to its receptor known by the skilled person, e.g. those described in the working examples.

As provided herein above, trafficking process of GB1a-containing receptors along the axon localizes intracellularly, and therefore a peptide needs to penetrate into a cell in order to bind to those receptors.

Thus, in one embodiment, the peptides of the present disclosure, as well as functional variants thereof, have membrane permeability (CP50) of 30 pM or less, such as 25 pM or less, such as 20 pM or less, such as 15 pM or less, such as 10 pM or less. Permeability of a peptide can be measured according to any conventional methods of for determining permeability of a peptide into a mammalian cell known by the skilled person, e.g. those described in the working examples.

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, have membrane permeability (CP50) of 28 pM or less, such as of 25 pM or less, such as of 20 pM or less, such as of 15 pM or less, such as of 10 pM or less, such as of 5 pM or less, such as about 1 pM.

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, have plasma stability (T1/2) of 12 hours or more, such as of 14 hours or more, such as of 16 hours or more, such as of 18 hours or more, such as of 20 hours or more, such as of 22 hours or more, such as of 24 hours or more, such as of 26 hours or more, such as of 28 hours or more, such as of 30 hours or more, such as of 32 hours or more, such as of 34 hours or more, such as of 38 hours or more, such as of 40 hours or more, such as of 42 hours or more, such as of 44 hours or more, such as of 46 hours or more, 48 hours or more.

Stability of a peptide, e.g. metabolic stability and/or plasma stability of a peptide, can be measured according to any conventional methods of for determining stability of a peptide in a mammalian system known by the skilled person, e.g. those described in the working examples.

In one embodiment, the peptides of the present disclosure, as well as functional variants thereof, cause an increase in the constitutive activity and a decrease in the ECso of GABA towards GB1a/2R, thus reducing the negative allosteric modulation effects of any one of AJAP-1 and/or PIANP at GB1a/2Rs.

Cyclic Peptides

In one embodiment, the peptide is cyclized to form a cyclic polypeptide. For example, a peptide may be cyclized by side chain-to-side chain, tail-to-side chain, side chain-to- head and head-to-tail. It will be evident to the skilled person that the peptides of the present disclosure can be made cyclic by any method known in the art. Common cyclization strategies include, but are not limited to, disulfide bridge between two cysteines (side chain-to-side chain), thioether bridge with e.g. a bromoacetic addition on the N-terminus and a cysteine (head-to-side chain) and lactamization either using coupling between a basic residue (Lys) and acid residues (Asp or Glu), or via native chemical ligation (NCL).

In one embodiment, the peptide is back-bone cyclized. In one embodiment, the peptide is “head-to-tail” cyclized, i.e. the N-terminal amino acid residue is linked to the C- terminal amino acid residue. In one embodiment, the N-terminal amino acid residue is linked to the C-terminal amino acid residue via a peptide bond. In one embodiment, the N-terminal amino acid residue is linked to the C-terminal amino acid residue via a nonpeptide chemical linker. Thus, in one embodiment, the peptides of the present disclosure further comprise: a. two cysteines linked via a non-peptide chemical linker; and/or b. a cysteine linked to the N-terminal amino acid residue of the peptide.

In one embodiment, the peptides of the present disclosure further comprise two cysteines.

In one embodiment, the peptides of the present disclosure further comprise a cysteine at the N-terminal. In one embodiment, the peptides of the present disclosure further comprise a cysteine at the C-terminal. In one embodiment, the peptides of the present disclosure further comprise a cysteine at the N-terminal and a cysteine at the C- terminal.

In one embodiment, a linker connects two cysteines within the peptide of the present disclosure.

In one embodiment, a linker connects a cysteine at the N-terminal amino acid residue and a cysteine at the C-terminal amino acid residue of the peptides of the present disclosure.

In one embodiment, the non-peptide chemical linker is:

Thus, in one embodiment, the peptide of the present disclosure is selected from the group consisting of:

NO: 480), wherein Xi to X11 are defined as in the section “Peptides” of the present disclosure.

Thus, in one embodiment, the peptide of the present disclosure is

(SEQ ID NO:

479), whereinX1 to X11 are defined as in the section “Peptides” of the present disclosure.

In one embodiment, the peptide of the present disclosure is

NO: 480) wherein Xi to X11 are defined as in the section “Peptides” of the present disclosure. Thus, in one embodiment, the peptide of the present disclosure is selected from the group consisting of: c. (SEQ ID NO: 477); and d. (SEQ ID NO: 478).

Thus, in one embodiment, the peptide of the present disclosure is selected from the group consisting of: . Thus, in one embodiment, the peptide of the present disclosure is selected from the group consisting of: and b. (SEQ ID NO: 478).

In one embodiment, the peptide of the present disclosure is

AC-CEDDDWIDWGPTSDDDC-NH 2 (SEQ ID NO: 475).

In one embodiment, the peptide of the present disclosure is In one embodiment, the peptide of the present disclosure is

In one embodiment, the peptide of the present disclosure is

Medicament/medical use

One aspect of the present disclosure relates to a peptide disclosed herein, as well as functional variants thereof, for use as a medicament.

The present inventors have found that the peptides of the present disclosure, as well as functional variants thereof, can act directly at presynaptic site and modulate the release of several excitatory neurotransmitters including glutamate and acetylcholine. This finds application in treatment of multiple diseases and disturbs.

Another aspect of the present disclosure relates to a peptide disclosed herein, as well as functional variants thereof, for use in the treatment of neurological and/or psychiatric disorders.

In one embodiment, the neurological disorder is epilepsy, spasticity, schizophrenia, dystonia, neuropathic pain, narcolepsy, spasticity, cognitive deficits such as Mild cognitive impairment (MCI), autism spectrum disorder and/or substance-use disorders.

In one embodiment, the neurological disorder is a cognitive deficit, for example MCI, in an individual suffering from Alzheimer’s disease.

In one embodiment, the psychiatric disorder is depression, anxiety and/or Attention deficit hyperactivity disorder (ADHD).

One aspect of the present disclosure relates to a method for treatment of neurological and/or psychiatric disorders, and symptoms thereof, comprising administration of a peptide according to the present disclosure, as well as functional variants thereof, to an individual in need thereof. One aspect of the present disclosure relates to a use of a peptide according to the present disclosure, as well as functional variants thereof, for the manufacture of a medicament for the treatment of neurological and/or psychiatric disorders, and symptoms thereof.

One aspect of the present disclosure relates to a medicament for use in the treatment or prevention of neurological and/or psychiatric disorders, and symptoms thereof comprising a peptide according to the present disclosure, as well as functional variants thereof.

Without being bound to theory, a peptide of the present disclosure, as well as a functional variant thereof, may be capable of increasing the release of dopamine and/or serotonin in an individual suffering from depressive disorders or ADHD.

Nucleic acid construct encoding a peptide

In one embodiment there is provided a nucleic acid construct encoding a peptide as defined herein. In one embodiment said nucleic acid construct will be able to continuously express said peptide for a prolonged period of time; such as at least 1 month, for example at least 2 months, such as at least 3 months, for example at least 4 months, such as at least 5 months, for example at least 6 months, such as at least 7 months, for example at least 8 months, such as at least 9 months, for example at least 12 months.

It is thus an aspect to provide a nucleic acid construct encoding a peptide as defined herewith.

In one embodiment there is provided a nucleic acid construct encoding a peptide of SEQ ID NO: 64, or a functional variant thereof, or any peptide disclosed herein, such as any peptide disclosed in any one of Table 3, table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15 and/or Table 16, or a functional variant thereof.

It is also an aspect to provide a nucleic acid construct encoding a peptide as defined herein for use in a method of treating neurological and/or psychiatric disorders as defined herein. By nucleic acid construct is understood a genetically engineered nucleic acid. The nucleic acid construct may be a non-replicating and linear nucleic acid, a circular expression vector or an autonomously replicating plasmid. A nucleic acid construct may comprise several elements such as, but not limited to genes or fragments of same, promoters, enhancers, terminators, poly-A tails, linkers, polylinkers, operative linkers, multiple cloning sites (MCS), markers, STOP codons, internal ribosomal entry sites (IRES) and host homologous sequences for integration or other defined elements. It is to be understood that the nucleic acid construct according to the present invention may comprise all or a subset of any combination of the above-mentioned elements.

Methods for engineering nucleic acid constructs are well known in the art (see, e.g., Molecular Cloning: A Laboratory Manual, Sambrook et al., eds., Cold Spring Harbor Laboratory, 2nd Edition, Cold Spring Harbor, N.Y., 1989). Further, nucleic acid constructs according to the present invention may be synthesized without template, and may be obtained from various commercial suppliers (e.g. Genscript Corporation).

In one embodiment, the nucleic acid constructs are naked DNA constructs comprising sequences encoding the triple agonist peptide.

Delivery vehicles

It is also an aspect to provide the nucleic acid construct as described herein above comprised within a delivery vehicle. A delivery vehicle is an entity whereby a nucleotide sequence or polypeptide or both can be transported from at least one media to another. Delivery vehicles are generally used for expression of the sequences encoded within the nucleic acid construct and/or for the intracellular delivery of the construct or the polypeptide encoded therein.

In one embodiment, there is provided a delivery vehicle comprising the nucleic acid construct as defined herein. A delivery vehicle may be selected from the group consisting of: RNA based vehicles, DNA based vehicles/ vectors, lipid based vehicles (such as a liposome), polymer based vehicles (such as a cationic polymer DNA carrier), colloidal gold particles (coating) and virally derived DNA or RNA vehicles or vectors. Methods of non-viral delivery include physical (carrier-free delivery) and chemical approaches (synthetic vector-based delivery).

Physical approaches, including needle injection, gene gun, jet injection, electroporation, ultrasound, and hydrodynamic delivery, employ a physical force that permeates the cell membrane and facilitates intracellular gene transfer. Said physical force may be electrical or mechanical.

Examples of chemical delivery vehicles include, but are not limited to: biodegradable polymer microspheres, lipid based formulations such as liposome carriers, cationically charged molecules such as liposomes, calcium salts or dendrimers, lipopolysaccharides, polypeptides and polysaccharides.

Recombinant cell

Another aspect of relates to a cell comprising the nucleic acid construct as defined herein. Such a recombinant cell can be used a tool for in vitro research, as a delivery vehicle for the nucleic acid construct or as part of a gene-therapy regime. The nucleic acid construct can be introduced into cells by techniques well known in the art which include microinjection of DNA into the nucleus of a cell, transfection, electroporation, lipofection/liposome fusion and particle bombardment. Suitable cells include autologous and non-autologous cells, and may include xenogenic cells.

Method of preparation (peptide)

In one aspect, the present disclosure relates to methods of manufacturing peptides according to SEQ ID NO: 64 or a functional variant thereof, or any peptide disclosed herein such as any peptide disclosed in any one of Table 3, table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, Table 12, Table 13, Table 14, Table 15 and/or Table 16, or a functional variant thereof.

Peptides according to the present invention may be prepared according to any conventional methods of peptide synthesis known by the skilled person, e.g. those described in the working examples. The starting materials for the processes described in the present application are known or may readily be prepared by conventional methods known by the skilled artisan from commercially available chemicals.

The peptides disclosed herein, as well as functional variants thereof, may be prepared by any methods known in the art; such as by standard peptide-preparation techniques including solution peptide synthesis or Merrifield-type solid phase peptide synthesis.

In one embodiment a peptide according to the disclosure is synthetically made or produced. The methods for synthetic production of peptides are well known in the art. Detailed descriptions as well as practical advice for producing synthetic peptides may be found in Synthetic Peptides: A User's Guide (Advances in Molecular Biology), Grant G. A. ed., Oxford University Press, 2002, or in: Pharmaceutical Formulation: Development of Peptides and Proteins, Frokjaer and Hovgaard eds., Taylor and Francis, 1999.

In one embodiment the peptide or peptide sequences of the disclosure are produced synthetically, in particular, by the sequence assisted peptide synthesis (SAPS) method, by solution synthesis, by solid-phase peptide synthesis (SPPS) such as Merrifield-type solid phase synthesis, by recombinant techniques (production by host cells comprising a first nucleic acid sequence encoding the peptide operably associated with a second nucleic acid capable of directing expression in said host cells) or enzymatic synthesis. These are well-known to the skilled person.

Peptides may be synthesised either batch-wise on a fully automated peptide synthesiser using 9-fluorenylmethyloxycarbonyl (Fmoc) or tert-Butyloxycarbonyl (Boc) as N-alpha-amino protecting group and suitable common protection groups for sidechain functionalities.

After purification such as by reversed phase HPLC, peptides may be further processed to obtain for example C- or N-terminal modified isoforms. The methods for terminal modification are well-known in the art.

Pharmaceutical formulations

In one aspect, the present invention relates to a composition comprising the peptide as disclosed herein. Preferably, said composition is pharmaceutically acceptable. In one embodiment the composition as described herein is in the form of a pharmaceutical formulation. In one embodiment, the composition as described herein further comprises a pharmaceutically acceptable carrier. In one aspect, the present invention concerns a composition comprising the compound as defined herein and a pharmaceutically acceptable carrier.

Items

1. A peptide comprising or consisting of the sequence

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

EX1 X2X3X4 XsXeW X7 X8 X9 X10 D X11 D (SEQ ID NO:64) wherein

Xi is Aspartic acid (D) or Threonine (T),

X2 is Aspartic acid (D) or Glutamic acid (E),

Xa is S, Aspartic acid (D), Tyrosine (Y) or Phenylalanine (F),

X4 is absent, Tryptophan (W), 3-(2-naphthyl)-L-alanine (2Nala), O-ethyl-L- tyrosine (Oetyr), O-benzyl-L-tyrosine (Obtyr), or L-Cysteic acid (Cya), X5 is Isoleucine (I) or Threonine (T),

Xe is Aspartic acid (D), Tryptophan (W) or Alanine (A),

X7 is Tyrosine (Y) or Glycine (G),

Xs is Glycine (G), Proline (P) or Aspartic acid (D),

X9 is Alanine (A), Threonine (T) or Serine (S),

Xw is absent, Glycine (G), Serine (S) or Alanine (A),

X11 is Aspartic acid (D), Threonine (T) or Glutamic acid (E), or a functional variant thereof, wherein the functional variant comprises 1 amino acid substitution.

2. The peptide according to item 1 , wherein said peptide is an inhibitor of GB1a/2Rs (heterodimeric gamma-amino butyric acid (GABA) B ia I GABAB2 receptor), such as wherein said peptide is an inhibitor of protein-protein interactions at GB1a/2Rs.

3. The peptide according to anyone of the preceding items, wherein: a. Xi is D; b. X 2 is D; c. X3 is S, D or Y, such as wherein X3 is D or Y, such as wherein X3 is D; d. X4 is absent, W, 2-Nal, O-Ey-Y or Obtyr; wherein X4 is absent; or wherein X4 is W, 2-Nal, O-Ey-Y or Obtyr, such as wherein X4 is W or 2- Nal; e. X5 is I; f. Xe is D; and/or g. Xs is P or D, or a functional variant thereof, wherein the functional variant comprises 1 amino acid substitution.

4. The peptide according to anyone of the preceding items, wherein X9 is A or S, and X10 is absent; or wherein X9 is T and X10 is G, S.

5. The peptide according to anyone of the preceding items, wherein X10 is G, S or absent, such as wherein X10 is G or S, such as wherein X10 is absent.

6. The peptide according to anyone of the preceding items, wherein X10 is absent and wherein X11 is D or T, such as wherein X10 is absent and wherein X11 is D; or wherein X11 is D or E and wherein X10 is G, S.

7. The peptide according to anyone of the preceding items, wherein the peptide is selected from the group consisting of:

SEQ ID NO: 15: TETEFIAWGPTGDED

SEQ ID NO: 16: EDDDWIDWYDSDDD

SEQ ID NO: 17: EDDDWIDWGPTSDDD

SEQ ID NO: 18: EDDYIDWYDSDDD

SEQ ID NO: 19: EDDYIDWGPTSDDD

SEQ ID NO: 20: EDDD(2Nala)IDWYDSDDD

SEQ ID NO: 21 : EDDD(2Nala)IDWGPTSDDD

SEQ ID NO: 22: EDDYTDWYDSDDD

SEQ ID NO: 23: EDDYTDWGPTSDDD

SEQ ID NO: 24: ETEFIAWGPTADED

SEQ ID NO: 25: ETEYIAWGPTGDED

SEQ ID NO: 26: ETEFIDWGPTGDED SEQ ID NO: 27: EDDSWIWWGPTGDED

SEQ ID NO: 28: EDDSWIWWYGADTD

SEQ ID NO: 29: EDDSWIDWGPTGDED

SEQ ID NO: 30: EDDSWIDWYGADTD

SEQ ID NO: 31 : ETEFI(Cya)WGPTGDED

SEQ ID NO: 32: EDDD(Oetyr)IDWYDSDDD

SEQ ID NO: 33: EDDD(Obtyr)IDWYDSDDD

SEQ ID NO: 34: EDDD(Otyr)IDWGPTSDDD

SEQ ID NO: 35: EDDD(Obtyr)IDWGPTSDDD or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions. The peptide according to anyone of the preceding items, wherein the peptide is N-terminally acetylated (Ac-N-ter) and/or C-terminally amidated (-NH2). The peptide according to anyone of the preceding items, wherein the peptide is conjugated to a cell-penetrating peptides (OPP), such as wherein the OPP is conjugated to the N-terminus or to the C-terminus of the peptide, optionally via a linker, such as via a peptidic linker. The peptide according to anyone of the preceding items, wherein the CPP is Tat, YGRKKRRQRRR (SEQ ID NO: 55),

R8, RRRRRRRR (SEQ ID NO: 56), R7, RRRRRRR (SEQ ID NO: 57), R6, RRRRRR (SEQ ID NO: 58), R5, RRRRR (SEQ ID NO: 59), R4, RRRR (SEQ ID NO: 60), R3, RRR, or R2, RR. The peptide according to anyone of the preceding items, wherein the peptide is selected from the group consisting of:

SEQ ID NO: 15: Ac-TETEFIAWGPTGDED-NH 2

SEQ ID NO: 16: Ac-EDDDWIDWYDSDDD-NH 2

SEQ ID NO: 17: Ac-EDDDWIDWGPTSDDD-NH 2 SEQ ID NO: 18: Ac-EDDYIDWYDSDDD-NH 2

SEQ ID NO: 19: Ac-EDDYIDWGPTSDDD-NH 2

SEQ ID NO: 20: Ac-EDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 21 : Ac-EDDD(2Nala)IDWGPTSDDD-NH 2

SEQ ID NO: 22: Ac-EDDYTDWYDSDDD-NH 2

SEQ ID NO: 23: Ac-EDDYTDWGPTSDDD-NH 2

SEQ ID NO: 24: Ac-ETEFIAWGPTADED-NH 2

SEQ ID NO: 25: Ac-ETEYIAWGPTGDED-NH 2

SEQ ID NO: 26: Ac-ETEFIDWGPTGDED-NH 2

SEQ ID NO: 27: Ac-EDDSWIWWGPTGDED-NH 2

SEQ ID NO: 28: Ac-EDDSWIWWYGADTD-NH 2

SEQ ID NO: 29: Ac-EDDSWIDWGPTGDED-NH 2

SEQ ID NO: 30: Ac-EDDSWIDWYGADTD-NH 2

SEQ ID NO: 31 : Ac-ETEFI(Cya)WGPTGDED-NH 2

SEQ ID NO: 32: Ac-EDDD(Oetyr)IDWYDSDDD-NH 2

SEQ ID NO: 33: Ac-EDDD(Obtyr)IDWYDSDDD-NH 2

SEQ ID NO: 34: Ac-EDDD(Oetyr)IDWGPTSDDD-NH 2

SEQ ID NO: 35: Ac-EDDD(Obtyr)IDWGPTSDDD-NH 2

SEQ ID NO: 36: Ac-RRRRRRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 37: Ac-RRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 38: Ac-RRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 39: Ac-RRRRRRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 40: Ac-RRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 41 : Ac-RRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 42: Ac-RRRREDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 43: Ac-RRRREDDD(2Nala)IDWGPTSDDD-NH 2 or a functional variant thereof, wherein the functional variant comprises 1 or 2 individual amino acid substitutions.

12. The peptide according to anyone of the preceding items, wherein the peptide has binding affinity (K) to a sushi domain 1/2 (SD1/2) protein of 20 nM or less; and/or wherein the peptide has membrane permeability (CP50) of 10 pM or less; and/or wherein the peptide has plasma stability (TI/ 2 ) of 12 hours or more. 13. The peptide according to anyone of the preceding items, wherein the peptide is capable of inhibiting an interaction between a protein and a SD1/2 of GB1a at presynaptic neurons; and/or wherein the peptide is capable of inhibiting an interaction between: sushi domain 1 (SD1) and adherence-junction associated protein 1 (AJAP1), SD1 and PILRa-associated neural protein (PIANP), and/or SD1 and amyloid precursor protein (APP).

14. A peptide according to anyone of the preceding items for use as a medicament.

15. A peptide according to anyone of items 1 to 13 for use in the treatment of neurological and/or psychiatric disorders, such as wherein the neurological disorder is epilepsy, spasticity, schizophrenia, dystonia, neuropathic pain, narcolepsy, spasticity, cognitive deficits such as Mild cognitive impairment (MCI), autism spectrum disorder and/or substance-use disorders ; and/or wherein the psychiatric disorder is depression, anxiety and/or Attention deficit hyperactivity disorder (ADHD).

Examples

Example 1. General methods and material.

Except where otherwise stated, amino acids and reagents for SPPS were purchased from Iris Biotech or Gyros Protein Technologies. Reagents and solvents were purchased from commercial suppliers and used without further purification.

Synthesized crude peptides were purified using a preparative reverse phase high performance liquid chromatography (RP-HPLC) system (Waters) with a reverse phase C18 column (Zorbax, 300 SB-C18, 21.2 x 250 mm) with a binary buffer system of H2O:CH3CN:TFA (A: 95:5:0.1; B: 5:95:0.1) at 20 mL/min. Mass spectra of peptides were characterized using the electron spray ionization (ESI) liquid chromatographymass spectrometry (LC-MS) coupled to an Agilent 6410 Triple Quadrupole Mass with a reverse phase C18 column (Zorbax Eclipse XBD-C18, 4.6 x 50 mm) using a binary buffer system consisting of H2O:CH3CN:formic acid (A: 95:5:0.1 ; B: 5:95:0.1) at 0.75 mL/min. The purity of synthesized peptides were determined by at 214 nm UV absorbance on an analytical reverse phase ultra-performance liquid chromatography (RP-UPLC) (Waters) system with a reverse phase C18 column (Acquity LIPLC BEH C18, 1.7 pm 2.1 x 50 mm) using a binary buffer system consisting of H2O:CH3CN:TFA (A: 95:5:0.1 ; B: 5:95:0.1) at 0.45 mL/min.

SPOT peptide array synthesis. pSPOT peptide arrays (CelluSpots, Intavis AG, Cologne, Germany) were synthesized using a RePepSL synthesizer (Intavis AG) on acid labile, amino functionalized, cellulose membrane discs (Intavis AG) containing 9- fluorenylmethyloxycarbonyl-p-alanine (Fmoc-p-Ala) linkers (minimum loading 1.0 pmol/cm). Synthesis was initiated by Fmoc deprotection using 20% piperidine in /V- methylpyrrolidone (NMP) (1 x 2 and 1 x 4 pL, 3 and 5 min, respectively) followed by washing with dimethylformamide (DMF, 7 x 100 pL per disc) and ethanol (EtOH, 3 x 300 pL per disc).

Peptide chain elongation was achieved using 1.2 pL of coupling solution consisting of preactivated amino acids (AAs, 0.5 M) with ethyl 2-cyano-2-(hydroxyimino)acetate oxyma (1.5 M) and /V,/V'-diisopropylcarbodiimide (DIC, 1.1 M) in NMP (2:1 :1 , aa:oxyma:DIC). The couplings were carried out 6 times (10, 15, 25, 30, 30 and 30 min, respectively), and subsequently, the membrane was capped twice with capping mixture (5% acidic anhydride in NMP), followed by washes with DMF (7 x 100 pL per disc). After chain elongation, final Fmoc deprotection was performed with 20% piperidine in NMP (3 x 4 pL, 5 min each), followed by 6x washes with DMF, subsequent N-terminal acetylation with capping mixture (3 x 4 pL, 5 min each) and final washes with DMF (7 x 100 pL per disc) and EtOH (7 x 200 pL per disc). Dried cellulose membrane discs were transferred to 96 deep-well blocks and were treated with the side-chain deprotection solution consisting of 80% TFA, 12% DCM, 5% H2O, and 3% TIPS (150 pL per well) for 1.5 h at room temperature (rt). Afterward, the deprotection solution was removed, and the discs were solubilized overnight at rt using a solvation mixture containing 88.5% TFA, 4% trifluoromethansufonic acid (TFMSA), 5% H2O, and 2.5% TIPS (250 pL per well). The resulting peptide- cellulose conjugates were precipitated with ice-cold ether (1 mL per well) and spun down at 1000 rpm for 90 min, followed by an additional wash of the formed pellet with ice-cold ether. The resulting pellets were redissolved in DMSO (500 pL per well) to give final stocks, which were transferred to a 384-well plate and printed (in duplicates) on white coated CelluSpots blank slides (76 x 26 mm, Intavis AG) using a SlideSpotter robot (Intavis AG).

Quality control of SPOT. Cleavable peptides as synthesis quality controls were prepared following general SPOT synthesis procedure (see above). For peptide cleavage from the cellulose support Fmoc-Rink amide linker was coupled as a first building block (4 times at10, 15, 25, and 30 min, respectively), followed by deprotection and standard SPOT peptide chain elongation procedure. The discs with synthesized peptides were transferred into Eppendorf tubes and treated with side chain deprotection solution 80% TFA, 12% DCM, 5% H2O, and 3% TIPS (150 pL per well) for 1.5 h. Subsequently, the cleaved peptides were precipitated with ice-cold diethyl ether and spun down at 14 000 x g (4 °C, 20 min). The precipitate was dissolved in 100 pL of 50:50:0.1 (CH3CN:H2O:TFA), filterted (Fisherbrand™ Syringe Filters: PTFE Membrane, 0.2 pM) and analyzed by LC-MS and RP- LIPLC.

SPOT peptide array screening based on slide imaging method. The DMSO stocks of SPOT synthesized peptide-cellulose conjugates were pipetted into a 384-well plate and then printed in duplicates onto the white coated CelluSpots slides (76 x 26 mm, Intavis AG) using a SlideSpotter robot (Intavis AG). After air dring overnight, the slides were rinsed gently with 1 x phosphate buffered saline (PBS, pH 7.4), and then blocked in blocking solution (5% bovine serum albumin (BSA) in PBS) for >2 h at rt on an orbital shaker. Subsequently, the slides were incubated with 6xHis-tagged SD1/2 protein in blocking solution at different concentrations (epitopes mapping: 0.25 pM, 0.01 pM and 0.05 pM for APP, AJAP-1 and PIANP, respectively. Deep mutational scans: 0.02 pM, 0.005 pM and 0.01 pM for APP, AJAP-1 and PIANP, respectively) for 1 h at rt. After briefly rinsing with washing solution (PBS-T: 0.05% Tween-20 in PBS buffer), the slides were washed 3 times by PBS-T for 2 min. Subsequently, slides were incubated with diluted HRP conjugated 6xHis Tag antibody (1 :10000, ab184607, abeam) for 30 min at rt. Finally, the slides were washed 5 x 2 min with washing solution and then visualized with SuperSignal West Femto Maximum Sensitivity Substrate (ThermoFisher Scientific) using Syngene PXi image system. The resulting SPOT slide images were analyzed using the Array Analyze Software (Active Motif) to quantify the relative SD1/2 binding of each peptide.

SPOT peptide array screening based on fluorescence polarization assay. FP assay was employed to explore the possibility of using SPOT synthesized peptide-cellulose conjugates to compete the binding between AJAP-1 pr obe (SEQ ID NO:15) and SD1/2 in solution to generate reproducible dose-response curves in black flat bottom 384-well plates (Corning Life Science). The DMSO stocks of all positional scan peptide -cellulose conjugates were diluted 10, 320 and 10 times (for APP, AJAP-1 , PIANP-derived peptides, respectively) directly into 1 xPBS (pH 7.4). All experiments were conducted following standard FP assay protocol by using 25 nM of AJAP-1 pr obe and 40 nM of recombinant SD1/2 protein (Dinamarca et al, 2019) in 100 mM PBS (pH 7.4) containing 1% bovine serum albumin (BSA) at rt. Subsequently, the fresh prepared diluted peptide-cellulose conjugates were pipetted into the flat bottom black 384-well plates (Corning Life Science) incubating with the premixed SDs/AJAP-1 probe. The fluorescence polarization values were measured using excitation/emission wavelengths of 530/580 nm with a Safire2 plate reader (Tecan). The instrumental Z-factor was adjusted to obtain the maximum fluorescence intensity and the G-factor was calibrated to give an initial 20 mP (millipolarization value) of the reference wells. The obtained the polarization value (mP) from each well represents the relative binding affinity of the mutated peptide compared to the corresponding wild type peptides.

Solution array synthesis.

Parallel synthesis in 96 format peptide synthesis was done using Intavis multipep RSi instrument. Fmoc-PAL resins was used (10 mg per well). Fmoc-amino acids (Fmoc- AA) were dissolved in dimethylformamide (DMF) containing 0.3 M OxymaPure. Throughout the synthesis Fmoc-AA were preactivated with diisopropylcarbodiimide (DIC) for 2 min before adding to the resin. Three couplings using 100 pL Fmoc-AA and 10 pL DIC (3 M in DMF) pr well were employed with coupling times of 5, 15 and 60 min. Removal of Fmoc was performed by washing twice with 120 pl per well of 25% piperidine in DMF for 2 and 8 min. Washing steps after deprotection with piperidine and after coupling steps included washing with DMF (150 pL per well) five times. After synthesis the resins were dried with ethanol and trifluoroacetic acid (TFA) was added using 92% TFA containing 4% triisopropylsilane (TIPS), 1 % thioanisol, and 3% H2O for 2 hours. This was done by adding 100 pL per well of the TFA cleavage cocktail over a period of 30 min adding up to a total volume of 1 mL to each well. TFA was then reduced in volume to approximately 100-150 pL by N2 flow and peptides were precipitated by the addition 800 pL of diethylether to each well. The peptides were transferred to Waters solvinert plates and washed thoroughly five times with diethylether 500 pL per well. After washing, the peptides were dried and then dissolved in 80% DMSO. Characterization was performed by UPLC-MS on a setup consisting of a Waters Acquity LIPLC system connected to an LCT Premier XE mass spectrometer from Micromass using solvent A 0.1% TFA in H2O and solvent B 0.1% TFA in CH3CN.

The peptides were confirmed by MS and had an average purity of >80%.

Peptide synthesis. All peptides were synthesized by solid-phase peptide synthesis (SPPS) using the Fmoc-strategy on the H-Rink amide ChemMatrix resin (Sigma- Aldrich) ranging from 0.01 to 0.1 mmol scale. Standard amide condensation reactions were carried out at rt under agitation using the coupling mixture with the equivalent ratio of 1 :4:4:8 (resin: protected AAs: HCTLI: DIPEA) and were monitored using a Kaiser Test Kit (Sigma-Aldrich). Fmoc deprotection was performed 2 times and 2 min for each using 20% piperidine in DMF and followed by extensive washing with DMF. After completion of elongation, the /V-terminal Fmoc was deprotected by 20% piperidine and then capped for 5 min using 5% acid anhydride in DMF at the presence of 8 equivalents of DI PEA and then the resin was washed with DMF and DCM before being dried under vacuum.

Automated peptide synthesis. Automated peptide synthesis using Fmoc-based SPPS was carried out either on a Prelude X or PurePep Chorus peptide synthesizers (Gyros Protein Technologies, Tucson, AZ, USA) with 10 mL (0.05 mmol scale) or 25 mL (0.1 mmol scale) glass reaction vessels using H-Rink amide ChemMatrix resin (Sigma- Aldrich).

For Prelude X, all reagents were prepared as DMF solutions: Fmoc-protected amino acids (0.2 M), HCTU (0.4 M), and DIPEA (1.0 M). Sequences were elongated as the following procedures: deprotection (2 x 2 min, rt, 300 rpm shaking) and coupling (2 x 5 min, 65 °C, 300 rpm shaking, for Cys and His couplings 2 x 5 min, 50 °C, 300 rpm shaking). Amino acids were double coupled using amino acid/HCTU/DIPEA (ratio = 1 : 1 :2.5) in 5-fold equivalent excess over the resin scale to ensure synthetic efficiency. For PurePep Chorus, all reagents were also prepared as solutions in DMF: Fmoc- protected amino acids (0.1 M), Oxyma Pure (0.4 M) and DIC (0.4 M). Sequences were elongated as the following procedures: deprotection (2 x 2 min, rt, 350 rpm shaking) and coupling (2 x 7 min, 65 °C, 350 rpm shaking, for Cys and His couplings 2 x 7 min, 50 °C, 350 rpm shaking). Amino acids were double coupled using amino acids/DIC/Oxyma pure (ratio = 1 :1.05:1.1) in 4-fold equivalent excess over the resin scale to ensure synthetic efficiency. TAMRA labelling. For FP assay, the fluorescent peptides were synthesized on resin by coupling the fluorophore TAMRA (Anaspec Inc.) onto the free /V-terminus of peptides. The reaction was performed at rt using a mixture of TAMRA/PyBOP/DIPEA (ratio = 1.5:1.5:3) in NMP. For minimizing photobleaching, the agitated reaction vessels were covered by tinfoil. After about 16 h, the resin was washed extensive with DMF and DCM followed by drying under vacuum before the final cleavage.

Peptide cleavage and purification. The synthesized peptides were cleaved from the resin using the cleavage cocktail containing 92.5% TFA, 2.5% H2O, 2.5% TIPS and 2.5% DODT for 2 h under agitation at rt. After cleavage, the peptides were precipitated with ice-cold diethyl ether and centrifuged at 4350 rpm for 10 min at 4 °C. The resulting crude peptides were redissolved in 50:50:0.1 (H2O: CH3CN: TFA), analyzed using LC- MS and LIPLC and lyophilized for purification (as described in General methods section). The collected fractions from purification were characterized by LC-MS. The purity of the fractions was determined at 214 nm using LIPLC. The fractions with higher than 95% purity were pooled and lyophilized for further experiments.

Fluorescence polarization (FP). FP assay was employed to determine the binding affinity of peptides in flat bottom black 384-well plates (Corning Life Science) using a Safire2 plate reader (Tecan). All experiments were conducted in 1xPBS (pH 7.4) containing 1% bovine serum albumin (BSA) at rt. The fluorescence polarization values were measured using excitation/emission wavelengths of 530/580 nm. The instrumental Z-factor was adjusted to obtain the maximum fluorescence intensity and the G-factor was calibrated to give an initial millipolarization value of 20 mP. All FP assays including saturation or competition experiments were performed using 25 nM of TAMRA labeled peptides as probes. For saturation assay, the millipolarization values were measured after mixing of probes with 12 serial 1 to 1 diluted concentrations of Sushi domains (SD1/2) protein. All experiments were performed in triplicate and the data were plotted to the one-site binding model using GraphPad Prism 9 to calculate the Kd of probes. In the competition assay, pre-mixed of SDs/probes were outcompeted with unlabeled peptides at 12 serial 1 to 1 diluted concentrations ranging from 0.24 to 500 pM based on the expected affinity. The experiments were performed at least in triplicate and the data were fitted to the sigmoidal dose response curve using GraphPad Prism 9 to obtain the IC50 of peptides. The Ki values were calculated according to Nikolovska-Coleska et al 310 . Results:

Taken together, SPOT identified several peptide hits from each APP, AJAP-1 and PIANP (data not shown), which were combined to three 21-mer length SDs binding epitopes (APP197-217, AJAP-1173-193 and PIANP93-113). Moreover, based on SPOT results the overlapping 9 residues length regions ( 203 EDDSDVWWG 211 SEQ ID NO: 4, 179 EFIAWGPTG 187 SEQ ID NO: 5, and "AIVWGPTVS 107 SEQ ID NO: 6) within APP, AJAP-1 and PIANP, respectively, were indicated representing the core binding regions to SD1/2. Additionally, alignment of all peptide hits to the previously reported two critical residues (WG) within the three proteins, the overlapped 5 amino acids, 207 DVWWG 211 SEQ ID NO: 7, 179 FIAWG 187 SEQ ID NO: 8 and "AIVWG 103 SEQ ID NO: 9 within APP, AJAP-1 and PIANP, respectively, were anticipated as the most essential residues for SD1/2 binding and highlighted in the red dashed frame.

High-affinity AJAP-1 pr obe facilitated determination of binding constants of peptides 11 (SEQ ID NO: 11) and 12 (SEQ ID NO: 12) (K = 5 and 1.3 pM, respectively). Thereafter, we successfully characterized the binding affinities of all synthesized SPOT hits by FP using the AJAP-1 pr obe (data not shown). AJAP-1 derived peptides exhibited the highest SD1/2 binding affinity with Ki values all below 100 nM. Additionally, peptide 15 (SEQ ID NO: 15) again proved to be the highest binding affinity hit (K = 23 nM). In contrast, peptides derived from the other two proteins exhibited much lower binding affinity, with peptides 13 (EDDSDVWWGGADTDY, SEQ ID NO: 13) (581 nM) and 14 (PWAIVWGPTVSREDG, SEQ ID NO: 14) (465 nM) being the best SPOT-identified hits from APP and PIANP, respectively (Table 3).

Example 2. Definition of the optimal binding motif

We first selected three peptides (peptides 15 (SEQ ID NO: 15), 36 (SEQ ID NO: 15) and 14 (SEQ ID NO: 14), displaying the highest binding affinity towards SD1/2, as a starting point for definition of optimal and the shortest possible binding interface. Thus, we next focused on systematic truncation scan at both N- and C-terminus of all three SD-binding peptides.

Results:

The information form elongation and truncation scans (Tables 4, 5 and 6) was used to design, synthesize and evaluate, the shortest possible 14-mer binding epitopes - APRWT (AC-EDDSDVWWGGADTD-NH 2 (SEQ ID NO: 1)), AJAP-1WT (AC- ETEFIAWGPTGDED-NH 2 (SEQ ID NO: 2)) and PIANPWT (Ac-PWAIVWGPTVSRED- NH 2 (SEQ ID NO: 3)). Calculated Ki values from FP experiments suggested a rank order of SD1/2 binding affinities - AJAP-1 (31 ± 1 nM) > > PIANP (476 ± 19 nM) > APP (557 ± 9 nM). Alignment of the three binding regions to the WG (positions 0 and 1, respectively) motif revealed overall similar structure consisting of a hydrophobic core (positions -3 to 2) surrounded by polar residues at both termini, apart from PIANPWT containing a more hydrophobic N-terminal.

Example 3. Alanine-scan, D-amino acid scan and N-methylation scan An Ala scan on the identified APPWT, AJAP-1 WT and PIANPWT was performed to elucidate the importance of each residue for the molecular recognition. 39 peptide variants (13 from each binding motif) each containing a single alanine substitution were synthesized and subsequently evaluated in the FP assay for their binding to SD1/2.

Similarly, a D-amino acid scan and /V-methylation scan were also conducted.

Results:

Comparison of the resulting binding affinities indicated similar tendencies as seen in truncation scan, underlining the importance of hydrophobic core (Figure 1). More specifically, the most critical residues were identified within the core at positions -2 and 0 (corresponding to positions 6 and 8 in the peptide consensus of claim 1 , SEQ ID NO: 64) and substitutions to Ala resulted in > 100-fold, whereas the Ala mutations at the remaining positions showed to affect the affinity to lesser extent. The major differences between different peptides were seen when substituting residues at positions 1-3. APPWT peptide Ala variants at these positions showed almost no changes in binding affinity, while AJAP-1 WT and PIANPWT Ala scan peptides exhibited 2-10-fold lower binding affinity to SD1/2.

Similarly, D-amino acids scan revealed that the stereochemistry of the hydrophobic residues in the core (positions -2, -1 and 0, corresponding to positions 6 to 8 in the peptide consensus of claim 1 , SEQ ID NO: 64) of peptides are critical for SD1/2 binding, resulting in peptides variants with 100-200-fold affinity decrease. Additionally, at none of the positions, incorporation of a D-amino acid resulted in an enhanced binding affinity of SD1/2. Similarly, to the previous scans, amino acid substitutions with the corresponding /V-Me amino acids again highlighted the importance of the core binding region. Specifically, /V-methylation at positions -1 and 1 (corresponding to positions 7 and 9 in the peptide consensus of claim 1 , SEQ ID NO: 64) exhibited the largest impact negative impact on the peptides binding to SD1/2 compared to the parent peptide variants. Interestingly, substitutions of position 2 amino acid (corresponding to position 10 in the peptide consensus of claim 1 , SEQ ID NO: 64) with /V-Me-Gly at APPwr yielded a peptide variant showing 3-fold increased affinity. Both AJAP-1WT and PIANPwr peptides at position 2 (corresponding to position 10 in the peptide consensus of claim 1, SEQ ID NO: 64) have a Pro residue, which provides rigidity. /V-Me-Gly substitution introduced in APPwr likewise introduce conformational constrains, thus potentially leading to preferred binding conformation and increased binding affinity.

The binding affinity for all synthesised and tested peptides are shown in Tables 7, 8 and 9.

Example 4. Deep mutational scan of APPwr, AJAP-1wr and PIANPwr peptides We applied the deep mutational scanning method using canonical amino acids (cAAs) for peptides of APPWT, AJAP-1WT and PIANPwr To achieve this, we again employed SPOT technology and generated three arrays and each comprising 280 14-mer peptides (including 14 wild peptides as references).

We synthesized additional positional scanning array (termed solution array) of APPWT using the conventional on-resin SPPS strategy in 96-well plates. This solution array contained 268 peptides (including two wild type peptides). Noteworthy, all peptides synthesized in this array showed around 90% crude purity. Taking advantage of the high purity of this peptide array, we generated six-point FP inhibition curve of each peptide and the obtained dose response curves enabled us to calculate the binding affinity (K) of each peptide variant (data not shown).

Results:

The obtained Ki (635 nM) using the crude APPWT (Table 10) in the solution array is almost identical to the previously measured K (557 nM) using the purified APPWT. The solution array results revealed dramatically increased affinity of the two most interesting mutants of APPWT, D207W and G211 Y, which showed Ki values of 78 nM and 167 nM, respectively. Substitution of D207 with bulkier hydrophobic amino acids including Phe, Tyr and Trp enabled additional contacts with hydrophobic residues at SD1.

FP-based screen of SPOT synthesized AJAP-1WT (Table 11) positional scan array identified 7 residues within AJAP-1WT, 179 EFIAWGP 185 (SEQ ID NO: 62), as the highly conserved core binding motif, which was consistent with the SPOT epitope mapping experiments. Multiple substitutions of the A182, located between two critical residues (1181 and W183), generated mutants exhibiting comparable or even higher binding affinity compared to AJAP-1wr. Particularly, the mutant A182D (peptide 26, SEQ ID NO: 26) represented the most promising mutation of AJAP-1WT with dramatically increased binding affinity (Ki = 9nM).

Two Arg residues are present in SD1 (R32 and R36) are in proximity to the A182.

Therefore, A182D mutation might facilitate additional electrostatic interaction between these thereby resulting in improved binding affinity.

T178 could be mutated into hydrophobic amino acids such as lie and Vai with improved affinity. Furthermore, F180Y, showed a potential to improve affinity.

Deep mutational scanning of PIANPwr (Table 12) revealed a highly conserved hydrophobic core binding motif composed of 6 residues ( 100 IVWGPT 105 (SEQ ID NO: 63)). Identified beneficial mutations of PIANPWT were relating mostly to mutations of hydrophobic residues into acidic amino acids. However, just slight affinity improvements (less than 2-fold) compared to the PIANPWT were observed.

Example 5. ncAA deep mutation scan of APPWT, AJAP-1wrand PIANPWT peptides We performed computational ncAAs based deep mutational scanning of APPWT, AJAP- 1WT and PIANPWT using the generated random forest (RF) models to predict the relative SD1/2 binding affinity of each mutant with introduced single ncAA. Specifically, each residue within peptides was individually replaced by any of the available 67 ncAAs in the z-scales database, resulting in 938 variants having a single ncAA introduced in each wild type peptide.

Results:

The beneficial ncAA mutations for APPWT were mainly related to mutation of residues Asp207 and Gly211 into aromatic and/or hydrophobic ncAAs. The relative affinities of peptides with ncAAs were organized as three individual heat maps (Figure 2). Based on the relative affinity, we selected and synthesized 17 peptides containing ncAAs and tested their binding affinity towards SD1/2 (Table 16). Overall, the computational prediction of the relative affinity showed a good correlation with the FP affinity data. Notably, all 7 mutations of the residue Asp207 within APPWT exhibited improved affinity (2- to 19-fold) compared to APPWT and 4 mutants exhibited even higher affinity (~ 2- to 4-fold) than the best cAA mutant Asp207Trp (K = 100 nM). Substitutions of Asp207 with 3-(2-naphthyl)-L-alanine (2-Nala) (K = 43 nM), O-benzyl-L-tyrosine (Obtyr) (K = 27.1 nM) and O-ethyl-L-tyrosine (Oetyr) (K = 46.3 nM) displayed 2-, 4- and 2-fold increased affinity compared to Asp207Trp, respectively. Furthermore, all 6 mutations of Gly211 also displayed greatly improved affinity (around 100 nM) compared to the wild type. For AJAP-1WT, the Ala182Cya (K = 11.8 nM) mutation showed similar affinity as Ala182Asp (K = 9), which represented the best identified cAAs mutant of AJAP-1WT, exhibiting 3-fold improved affinity relative to the wild type.

Example 6. Affinity maturation

Design of novel SD-specific high affinity peptides based on APPWT and AJAP-1WT. We first divided both wild type peptides into the N-terminal and C-terminal parts, separating them at positions 0 and 1. Then for the design of novel peptides one part (either N- or C-terminal) was taken from APPWT and other part (either N- or C-terminal) from AJAP-1WT, generating two hybrid peptides X (AC-EDDSDVWWGPTGDED-NH2 SEQ ID NO: 48) and Y (Ac-ETEFIAWGGADTD-NH 2 SEQ ID NO: 49). The hybrid peptide X was obtained by merging APPWT N-terminal part and AJAP-1WT C-terminal part and displayed similar affinity as AJAP-1wr that is almost 10-fold higher than APPWT. In contrast, the hybrid peptide Y containing AJAP-1WT N-terminal part and C- terminal part of APPWT exhibited comparable affinity as APPWT (Table 1). Next, to further optimize binding properties we introduced the best C-terminal mutation G211Y of APPWT into peptide Y generating a chameleon peptide 37 (Ac-ETEFIAWYGADTD- NH2 SEQ ID NO: 50), which showed improved affinity comparable to peptides X and AJAP-1WT (Table 2). Then, we substituted the Pro in the AJAP-1WT, generating peptide 38 (Ac-ETEFIAWG(/V-Me-G)TGDED-NH 2 SEQ ID NO: 51), which exhibited the same affinity as AJAP-1WT indicating that the /V-Me-Gly can mimic peptide binding conformation introduced by Pro (Table 1). After exploring contributions of single modifications, we moved forward with incorporation of multiple beneficial mutations into native or hybrid peptide variants. First, we investigated the effect of introducing double modification, namely D207W and V208I into peptide X.

This led to peptide 27 (Ac-EDDSWIWWGPTGDED-NH 2 SEQ ID NO: 27) showing 3.8- fold better affinity than 9. With this information we combined D207W and V208I with G211Y mutations and incorporated them to APPwr, resulting peptide 28 (Ac- EDDSWIWWYGADTD-NH2 SEQ ID NO: 28) displayed K\ value almost identical to peptide X (Table 1). Encouraged by synergistic effect of multiple mutations on the peptides binding affinity we in introduced the best mutation (A182D) of AJAP-1WT into peptides 27 and 28 at the corresponding positions, generating peptides 29 and 30 with further improved affinity (Table 1). Additionally, we introduced other identified favourable mutations of APPWT including S206D, G212D, T215D and A213S into peptide 30. Additionally, affinity improving mutations (G187S and E189D) from AJAP- 1wr SAR study were introduced into the AJAP-1wr derived C-terminal part of peptide 29, together with S206D. Satisfyingly, all incorporated mutations into peptides APPWT and X were compatible and generated peptides 16 (AC-EDDDWIDWYDSDDD-NH2 SEQ ID NO: 16) and 17 (Ac-EDDDWIDWGPTSDDD-NH 2 SEQ ID NO: 17) with low nanomolar affinity. Similarly, optimized versions of AJAP-1wr and peptide Y, resulted in two high affinity peptides 18 (Ac-EDDYIDWGPTSDDD-NH 2 SEQ ID NO: 18) and 19 (AC-EDDYIDWYDSDDD-NH2 SEQ ID NO: 19) (Table 1).

The 3 the most promising hydrophobic ncAAs were Obtyr, 2Nala and Oetyr, which showed the greatest improvement to the affinity of APPWT by substituting the D207 residue. We incorporated these ncAAs into corresponding positions within peptides 16 and 17. The FP data showed that all but one of resulting peptides (peptides 32-35, SEQ ID NOs: 36 to 39) exhibited slightly higher affinity compared to peptides 16 and 17. In contrast, introduction of the 2Nala into peptide 16 further increased affinity to 1.5 nM (peptide 20). (Table 1). The full list of the synthesised and tested peptides for affinity maturation can be found in Table 13.

Table 1. Overview of synthesized peptides for affinity maturation. Data are displayed as calculated Ki values (mean ± SEM, n > 3). All peptides have N-terminal acetylation and C-terminal amidation.

Example 7 Pull-down experiments

The aim of these experiments was to examine whether the most potent peptide 20 could bind to native SD and examine the selectivity towards the SD.

Covalent immobilization of peptides. Peptides AJAP-1WT, 17 and 20 comprising a C-terminal Cys and a PEG2 linker, were loaded covalently to Dynabeads M-270 Epoxy beads (Thermo Fischer Scientific) according to the protocol of the manufacturer. 25 pL DMSO stocks of peptides (10 mM) were diluted 15 times into 350 pL coupling buffer (100 mM PBS buffer containing 2 M (NH4)SO4 and 10 mM TCEP, pH = 7.4) and agitated overnight at 37 °C with Dynabeads M-270 Epoxy beads (5 mg, ~3.3 x 10 8 beads), which had been washed and equilibrated with coupling buffer before. After removing excess peptides, the Dynabeads M-270 Epoxy beads were washed three times and equilibrated with 100 mM PBS buffer, pH = 7.4 and stored at 4 °C.

Preparation of whole-brain lysates. Dissected adult mouse brains were placed in ice- cold homogenization buffer (320 mM sucrose, 4 mM HEPES, pH 7.5, 1 mM EDTA, 1 mM EGTA, supplemented with complete EDTA-free protease inhibitor mixture (Roche), 1 mL per 100 mg of brain) and homogenized with a glass-Teflon homogenizer (40 passes) on ice. The homogenized material was cleared by centrifugation at 500 * g (4 °C, 15 min). The supernatant was carefully removed and ultracentrifuged at 48 000 x g (4 °C, 45 min) to isolate membrane-enriched fraction (pallet), which was solubilized for 3 h at 4 °C in NETN buffer (100 mM NaCI, 1 mM EDTA, 0.5% Nonidet P-40, 20 mM Tris-HCI, pH 7.4, supplemented with a protease inhibitor mixture). The solubilized membrane-enriched fraction was cleared by ultracentrifugation at 100 000 x g (4 °C, 45 min). The final brain lysates (at 2 mg/mL protein concentration) were directly used for immunoblot analysis (input lanes) or for pull-down experiments.

Pull-down experiments. The covalently immobilized peptides AJAP-1WT, 16 and 20 were washed with 3 x 1 mL of 100 mM PBS and 1 mL of 100 mM PBS containing 0.1% Tween-20 and directly incubated with freshly prepared whole-brain lysate (2 mg/mL of total protein) overnight on a spring wheel at 4 °C. The next day, the beads were washed with 2 x 1 mL of 100 mM PBS and 2 x 1 mL of 100 mM PBS containing 0.1% Tween-20, and then the pulled down proteins were eluted with 4 x sample loading buffer containing 200 mM DTT (2 x 10 pL). Proteins were resolved using standard onedimensional SDS-PAGE on 10% polyacrylamide gels (for 45 min at 70 mV, followed by an additional 1.5 h at 120 mV). For immunoblotting analysis, proteins were transferred to nitrocellulose membrane (0.2-pm pore size) (iBIot™ 2 Transfer Stacks, nitrocellulose, mini) using i Blot2 (invitrogen) and then probed with the primary antibodies mouse anti-SD clone 43N12 (1:250) or mouse monoclonal to GABABI subunit (abeam, 1 :500) in combination with secondary peroxidase-coupled sheep antimouse IgG antibody (Cytiva, 1:5000).

Results:

The pull-down experiments demonstrated that both AJAP-1WT and peptide 20 showed the ability to bind and enrich full-length GB1a subunit in a complex native environment, whereas the negative control peptide Scr-20 and beads alone did not interact with the GB1a subunit (Figure 3).

Example 8. In vitro plasma stability.

For time-course assay, 360 pL human plasma was pre-warmed up at 37 °C for 15 min and then spiked with 40 pL of tested peptides (1 mM in 1 x PBS). After 0, 2, 6, 12, 24 and 48 h incubation at 37 °C, for each time point 45 pL sample was collected and pre-treated with 50 pL 6 M urea for 10 min at 0 °C. Subsequently, 100 pL acetonitrile was added into and then samples were incubated overnight at -20 °C. After that, all samples were centrifuged at 13,400 rpm for 10 min and the supernatant was filtered and then analyzed by LC-MS and LIPLC (Waters). The area under the curve (AUG) was determined and normalized to the value of the first time point. The half-life (7I/ 2 ) was obtained by fitting the data to the one-phase decay equation in GraphPad Prism 9 and expressed as the mean of three individual experiments. For end-point assay, 108 pL human plasma was pre-warmed up at 37 °C for 15 min and then spiked with 12 pL of tested peptides (1 mM in 1 x PBS). After 0 and 48 h incubation at 37 °C, 45 pL sample was collected and then treated as following the same procedure described in time-course assay above. The area under the curve (AUG) of end-point (48 h incubation) was normalized to the value of the first time point and then the stability was expressed as compound remaining (%).

Results:

It was found that even the native protein-derived wild type peptides (APPWT, AJAP-1WT and PIANPWT) exhibited excellent stability in human plasma with half-life times of >48, >48 and 16 h, respectively. Similarly, the half life of all four optimized peptides composed of cAAs (16-19) were >48 hours and no apparent degradation was observed after two days incubation with 90% of intact peptide remaining. Moreover, we also investigated the impact of polyR tagging on the plasma stability of peptides. The results demonstrated that R8-tagged peptides (R8-16 and R8-17) showed excellent stability as well with about 50% remaining after 48 hours incubation. Interestingly, truncation of the poly-R from 8R to 3R further improved plasma stability (R3-16 and R3-17), and the amount of the remaining peptide increased from 50% to about 80% after 48 hours incubation (Figure 4).

Additionally, we evaluated the influence of incorporating the ncAA (2Nala) into 4R tagged version of peptides by characterization of their affinity, permeability, and plasma stability. We found that 4R-20 exhibited about 2-fold higher affinity than 4R-16, while 4R-21 showed almost identical affinity as 4R-17 (Figure 4). Moreover, the two 4R tagged peptides containing 2Nala (4R-20 and 4R-21) displayed excellent plasma stability (>48 hours half-life) and cell membrane permeability (~1 pM CP50) (Figure 4 and Table 2). In conclusion, it was demonstrated that truncating the polyR moieties of our CPP tagged peptides to 4R resulted in excellent membrane permeability, while displaying much less negative impact on the affinity, exhibiting higher human plasma stability compared to 8R tagged peptides.

The full list of the synthesised and tested peptides in combination with a CPP can be found in Table 14.

Example 9. Chloroalkane penetration assay (CAPA).

The HeLa cell line used for CAPA assay was generated by Chenoweth and co-workers and provided by Kritzer and co-workers. The cells stably express a HaloTag-GFP fusion protein anchored through a mitochondria targeting peptide exposed to the cytosol. For CAPA, HeLa cells were seeded using DM EM supplemented with FBS in a 96-well plate one day before the experiment at a density of 4 x 10 4 cells/well. Just before the assay, the growth media was aspirated off and replaced with Opti-MEM assay media (100 pL/well). DMSO stocks of peptides (10 mM) were diluted in Opti- MEM to prepare serial dilutions of peptides in a separate 96-well PCR plate ensuring the final concentration of DMSO was kept consistent and below 1%. Subsequently, 25 pL of peptide solution was added into each well and the plate was incubated for 4 h at 37 °C with 5% CO2. After aspirating the assay media off, cells were washed with fresh Opti-MEM (80 pL/well) for 15 min at rt. Following removal of the washing media, cells were incubated with 5 pM CA-TAMRA in Opti-MEM (50 pL/well) for 15 min. Non CATAMRA control wells were incubated with Opti-MEM alone (50 pL/well). After removal of the dye containing media, cells were washed with fresh Opti-MEM (80 pL/well) for 30 min. After aspiration, cells were trypsinized using clear Trypsin (20 pL/well, 10 min at 37 °C), resuspended in PBS containing 2% FBS (180 pL/well), and analyzed using a benchtop flow cytometer (Guava EasyCyte, EMD Millipore).

Results:

Due to high similarity, we chose peptides 16 and 17 as models for characterizing and potential optimizing their cell membrane permeability. As expected, CAPA results of CA-tagged peptides 16 and 20 showed only poor cell membrane permeability with CP50 values around 150 pM indicating that it is difficult for highly negatively charged peptides to penetrate cell membranes. Strikingly, the other two similar peptides 17 and 21 showed 2- to 7-fold better membrane permeability compared to peptides 16 and 20, with 17 being the best in permeating the cell membrane (CP50 = 21 pM) (Table 2). The full list of the synthesised and tested CA-tagged peptides can be found in Table 15.

Table 0 Overview of sythesized CPP tagged peptides. Values of calculated SD1/2 binding affinity (K) , cell membrane permeability (CP50) and plasma stability (7I/ 2 ) are expressed as mean ± SEM, n > 3.

To improve cell membrane permeability of the peptides 16 and 17, we decided to conjugate the two prototypical CPPs, Tat (YGRKKRRQRRR SEQ ID NO: 28) and polyR (SEQ ID NOs: 29 to 33) to the N-terminus. The CAPA results demonstrated that both Tat and R8 N-terminally attached to 16 and 17 (R8-16, Tat-16, R8-17 and Tat-17) significantly improved the conjugates permeability with CP50 values around 1 pM (Table 2). However, we found that the positively charged CPPs dramatically influenced the affinity of the negatively charged SD1/2 binding peptides 16 and 17 resulting in 12- to 33-fold loss of affinity. changing the position of CPPs from the N- to C-terminus had almost no effect of the affinity of the peptides. Moreover, changing in the CPP position resulted in 3- to 23-fold higher CP50 values compared to the corresponding N-terminally tagged peptides (Table 2).

Comparing to CPP-tagged peptides without linkers, adding linkers between CPPs and peptides did not improve binding affinity but in contrast the linker-containing peptides exhibited lower affinity (R8-FL-16/17 and R8-RL-16/17 versus R8-16/17, respectively) (Table 2). The linkers tested were a rigid linker (RL) of sequence EAAAK (SEQ ID NO: 60) and a more flexible linker (FL) of sequence GGGGS (SEQ ID NO: 61).

The CAPA results revealed that truncation to 4R still maintained excellent cell membrane permeability similar to the R8-tagged version with a CP50 around 1 pM. However, further reduction to 3R resulted in substantially decreased permeability (CP50 = ~10 pM) (Figure 4).

Example 10. SRE-luciferase accumulation assay.

To elucidate whether our developed peptides AJAP-1WT, 16 and 20 could interact and disrupt the trans-interaction between AJAP-1 and GB1a/2Rs, we used an established accumulation assay based on coupling GB1a/2Rs to phospholipase C (PLC) via chimeric Ga q j (Conklin et al. 1993). PLC activity is then monitored with a serum responsive element-luciferase (SRE-Luciferase) reporter amplifying the GB1a/2Rs response (Yoo et al. 2017) (Figure 5).

HEK293T cells stably expressing Ga q /j were transiently transfected with Flag-GB1a, Flag-GB2 and SRE-FLuc with or without AJAP-1. In order to ensure GB1a/2 binding of AJAP-1 in trans, a pool of HEK293T-Ga q /j cells expressing AJAP-1 was mixed with HEK293-Ga q /j cells expressing Flag-GB1a, Flag-GB2 and SRE-FLuc. Transfected cells were distributed into 96-well microplates (Greiner Bio-One) at a density of 80,000 cells/well. After 24 h, the culture medium was replaced with Opti-MEM™-GlutaMAX™. Peptides were incubated in Opti-MEM™-GlutaMAX™ for 10 minutes. In presence of peptide, GB1a/2 receptors were activated with various concentrations of GABA for 6 h. FLuc activity in lysed cells was measured using the Luciferase® Assay Kit (Promega) and a Spark® microplate reader. Luminescence signals were adjusted by subtracting the luminescence obtained when expressing SRE-FLuc fusion proteins alone.

Results:

The presence of AJAP-1 protein in trans significantly reduced the constitutive activity of transcellular GB1a/2Rs and shifted the ECso towards a higher GABA concentration without affecting the maximum efficacy (E ma x) (Figure 5). The addition of 10 pM AJAP- 1wr, 16 or 20 in presence of AJAP-1 elevated basal activity of GB1a/2Rs (Figure 5) and decreased ECso values (Figure 5) of GABA activation at trans-cellular GB1a/2Rs to similar levels as receptors in absence of AJAP-1 . Noteworthy, the developed low nanomolar affinity peptides (16 and 20) reduced the negative allosteric modulation effects of AJAP-1 at GB1a/2Rs in trans to a greater extent than AJAP-1WT peptide, highlighting the importance of peptide optimization (Figure 5). In comparison, when treated with a 10 pM solution of the scrambled control peptide Scr-20 in the presence of AJAP-1 , trans-cellular GB1a/2Rs still showed significant decreased basal activity (Figure 5) and increased ECso values of GABA at GB1a/2Rs (Figure 5). Overall, all peptides (AJAP-1 wr, 16, 20 and Scr-20) did not affect the E ma x values of GABA- activated GB1a/2Rs in the absence of AJAP-1 (Figure 5).

Peptides AJAP-1WT, 16 and 20 specifically disrupt the transcellular AJAP-1/GB1a/2Rs interaction and reduce the negative allosteric modulation effects of AJAP-1 at GB1a/2Rs, and thus modulate the pharmacological profiles of GABA at GB1a/2Rs.

Example 11. Synthesis of cyclic peptides.

Linear peptides comprising two cysteine residues were solubilized in a solution consisting of 50% acetonitrile (ACN) and NH4CO3 buffer (pH=8.5) at a concentration of 1 mM. Subsequently, the linker, Xylylene dibromide, for cysteine cross-linking was dissolved in ACN at a concentration of 12 mM. The ACN solution of Xylylene dibromide was added dropwise into the peptide solution at room temperature under vigorous stirring until it reached a 1.2-fold excess relative to the quantity of peptides.

For linear peptides containing a single cysteine and chloroacetic acid-capped N- terminus, the cyclization reaction was initiated by dissolving these peptides in a solution comprising 50% ACN and NH4CO3 buffer (pH=8.5) at a concentration of 1 mM. Subsequently, the conversion of linear peptides into cyclic peptides was monitored using Ultra-Performance Liquid Chromatography (UPLC) and Liquid Chromatography- Mass Spectrometry (LC-MS). Upon completion of cyclization, the crude cyclized peptides were purified through Reversed-Phase High-Performance Liquid Chromatography (RP-HPLC). Cyclic peptides generated are disclosed in table 17 of the present disclosure.

Sequence overview

SEQ ID NO: 4: EDDSDVWWG, is APP(203-211)

SEQ ID NO: 5: EFIAWGPTG, is AJAP-1 (179-187)

SEQ ID NO: 6: AIVWGPTVS, is PIANP(99-107)

SEQ ID NO: 7: DVWWG, is APP(207-211)

SEQ ID NO: 8: FIAWG, is AJAP-1 (180-184)

SEQ ID NO: 9: is AIVWG, is PIANP(99-103)

SEQ ID NO: 28: YGRKKRRQRRR is cell-penetrating peptide Tat

SEQ ID NO: 29: RRRRRRRR is cell-penetrating peptide polyR (R8)

SEQ ID NO: 30: RRRRRRR is cell-penetrating peptide polyR (R7)

SEQ ID NO: 31 : RRRRRR is cell-penetrating peptide polyR (R6)

SEQ ID NO: 32: RRRRR is cell-penetrating peptide polyR (R5)

SEQ ID NO: 33: RRRR is cell-penetrating peptide polyR (R4)

SEQ ID NO: 60: Rigid Linker (RL) EAAAK

SEQ ID NO: 61 : Flexible Linker (FL): GGGGS

SEQ ID NO: 62: EFIAWGP

SEQ ID NO: 63: IVWGPT

EX1 X2 X3X4 XsXe W X7 X8 X9 X10 D X11 D (SEQ ID NO:64)

All peptides listed in any one of Table 3 to table 16 are N-terminally acetylated and C- terminally amidated.

Table 3. SPOT hit (epitopes mapping) validation and previously reported peptides PWAIVWGPTVSREDG 465 ± 16 75

WAIVWGPTVSREDGG 1347 ± 49 76

AIVWGPTVSREDGGD 637 ± 45 77

Table 4. Truncation and elongation scans of APP binding epitope EEDDSDVWW 1786 ± 16 107

EEDDSDVW >62500 108

Table 5. Elongation and Truncation scan of AJAP-1 binding epitope

Table 6. Elongation and Truncation scan of PIANP binding epitope QYPWAIVWGPTVSREDG 650 ±9 132

YPWAIVWGPTVSREDG 433 ±2 133

PWAIVWGPTVSREDG 463 ± 10 134

PWAIVWGPTVSREDGG 561 ±21 135

PWAIVWGPTVSREDGGD 716 ±6 136

PWAIVWGPTVSREDGGDP 597 ±7 137

PWAIVWGPTVSREDGGDPN 579 ±21 138

PWAIVWGPTVSREDGGDPNS 706 ± 16 139

VWGPTVSREDG 1740 ±62 140

IVWGPTVSREDG 15700 ±210 141

AIVWGPTVSREDG 5550 ± 11 142

WAIVWGPTVSREDG >62500 143

PWAIVWGPTVSREDG 29900 ± 2000 144

PWAIVWGPTVSRED 762 ± 13 145

PWAIVWGPTVSRE 650 ±9 146

PWAIVWGPTVSR 433 ±2 147

PWAIVWGPTVS 463 ± 10 148

PWAIVWGPTV 561 ±21 149

PWAIVWGPT 716 ±6 150

PWAIVWGP 597 ±7 151

PWAIVWG 579 ±21 152

PWAIVW 706 ± 16 153

Table 7. Ala-, D-amino acids and /V-methylation scans of APPWT Table 8. Ala-, D-amino acids and /V-methylation scans of AJAP-1WT

Table 9. Ala-, D-amino acids and /V-methylation scans of PIANPwr PWAIVWGPtVSRED 1300 ± 10 257

PWAIVWGPTvSRED 686 ± 19 258

PWAIVWGPTVSRED 552 ± 4 259

PWAIVWGPTVSrED 605 ± 15 260

PWAIVWGPTVSReD 760 ± 25 261

PWAIVWGPTVSREd 725 ± 3 262

P(/V-Me-W)AIVWGPTVSRED 833 ± 12 263

PW(/V-Me-A)IVWGPTVSRED 7233 ± 130 264

PWA(/V-Me-I)VWGPTVSRED 370 ± 5 265

PWAI(/V-Me-V)WGPTVSRED 6999 ± 100 266

PWAIV(/V-Me-W)GPTVSRED 636 ± 6 267

PWAIVW(/V-Me-G)PTVSRED 58309 ± 4414 268

PWAIVWG(/V-Me-G)TVSRED 525 ± 22 269

PWAIVWGP(/V-Me-T)VSRED 1265 ± 27 270

PWAIVWGPT(/V-Me-V)SRED 1778 ± 87 271

PWAIVWGPTV(/V-Me-S)RED 1045 ± 1 272

PWAIVWGPTVS(/V-Me-R)ED 502 ± 15 273

PWAIVWGPTVSR(/V-Me-E)D 523 ± 13 274

PWAIVWGPTVSRE(/V-Me-D) 434 ± 10 275

Table 10. Deep mutational scanning validation variants of APPWT EDDSDVWWHGADTD 270 ± 4 287

EDDSDVWWYGADTD 108 ± 1 288

EDDSDVWWGPADTD 332 ± 13 289

EDDSDVWWGGHDTD 346 ± 6 290

EDDSDVWWGGSDTD 204 ± 4 291

EDDSDVWWGGYDTD 255 ± 11 292

EDDSDVWWGGADED 337 ± 10 293

EDDSDVWWGGADHD 739 ± 9 294

EDDSDVWWGGADWD 459 ± 21 295

Table 11. Deep mutational scanning validation variants of AJAP-1WT

Table 12. Deep mutational scanning validation variants of PIANPwr PWVIVWGPTVSRED 302 ±4 348

PWYIVWGPTVSRED 336 ±2 349

PWFIVWGPTVSDED 192 ±4 350

PWAIDWGPTVSRED 1302 ± 16 351

PWAIVWGPTSSRED 383 ±3 352

PWAIVWGPTDSRED 417 ±6 353

PWAIVWGPTVDRED 379 ±9 354

PWAIVWGPTVSDED 327 ±6 355

AIVWGPTVSRED 683 ± 41 356

AIVWGPTVSRED* 773 ± 11 357

FIVWGPTVSRED 367 ± 18 358

FIVWGPTVSRED* 503 ±9 359

Table 13. Peptides for affinity maturation EDDSWIDWYGADTD 11 ±0.1 379

EDDSWIDWGPTGDED 5.6 ±0.7 380

EDDDWIDWYDSDDD 3.2 ±0.2 381

EDDDWIDWGPTSDDD 4.0 ±0.3 382

EDDYIDWYDSDDD 4.6 ±0.3 383

EDDYIDWGPTSDDD 3.8 ±0.2 384

Table 14. CPPs tagged peptides RRRRRREDDDWIDWGPTSDDD 43.4 ± 1 409

RRRRREDDDWIDWGPTSDDD 32 ± 1 410

RRRREDDDWIDWGPTSDDD 15 ± 1 411

RRREDDDWIDWGPTSDDD 9 ± 0.2 412

RRRREDDD(2Nala)IDWYDSDDD 15 ± 1 413

RRRREDDD(2Nala)IDWGPTSDDD 13 ± 1 414

RRRR(2Nala)EDSDDYDDIDDDW 142537 ± 2504 415

Table 15. CA-taqqed peptides for CAPA

Kt (nM) SEQ ID NO:

Sequence

CA-ETEFIAWGPTGDED 45 ± 4 416 CA-EDDDWI DWYDSDDD 153 ± 21 417 CA-EDDDWIDWGPTSDDD 21 ± 2 418 CA-EDDD(2Nala)IDWYDSDDD 146 ± 7 419 CA-EDDD(2Nala)IDWGPTSDDD 72 ± 6 420 CA-(2Nala)EDSDDYDDIDDDW 118 ± 15 421 CA-RRRR(2Nala)EDSDDYDDIDDDW 0.6 ± 0.2 422 CA-RRRREDDD(2Nala)IDWYDSDDD 1.1 ± 0.2 423 CA-RRRREDDD(2Nala)IDWGPTSDDD 1.2 ± 0.3 424 CA-RRRRRRRREDDDWIDWGPTSDDD 1.3 ± 0.7 425 CA-RRRRRRRREDDDWIDWYDSDDD 1.5 ± 0.9 426 CA-EDDDWIDWGPTSDDDRRRRRRRR 17 ± 1.6 427 CA-EDDDWIDWYDSDDDRRRRRRRR 17 ± 0.2 428

CA-YGRKKRRQRRREDDDWIDWGPTSDDD 2.4 ± 1.0 429 CA-YGRKKRRQRRREDDDWI DWYDSDDD 0.8 ± 0.1 430 CA-EDDDWI DWYDSDDDYGRKKRRQRRR 19 ± 0.4 431 CA-EDDDWI DWGPTSDDDYGRKKRRQRRR 6.1 ± 0.8 432 CA-EDDDWI DWYDSDDDEAAAKRRRRRRRR 10 ± 1.5 433 CA-EDDDWIDWYDSDDDGGGGSRRRRRRRR 9.0 ± 1.2 434 CA-RRRRRRRREDDDWIDWYDSDDD 1.5 ± 0.9 435 CA-RRRRRRREDDDWIDWYDSDDD 0.6 ± 0.4 436 CA-RRRRRREDDDWIDWYDSDDD 0.5 ± 0.3 437 CA-RRRRREDDDWIDWYDSDDD 0.5 ± 0.2 438 CA-RRRREDDDWIDWYDSDDD 1.0 ± 0.4 439

Table 16. ncAAs incorporated peptides

SEQ ID NO: 1 : Ac-EDDSDVWWGGADTD-NH 2 , is (APPWT)

SEQ ID NO: 2: Ac-ETEFIAWGPTGDED-NH 2 , is (AJAP-1WT)

SEQ ID NO: 3: AC-PWAIVWGPTVSRED-NH 2 , is (PIANPWT)

SEQ ID NO: 10: TAMRA-TETEFIAWGPTGDED is AJAP-1p robe

SEQ ID NO: 11 : DSADAEEDDSDVWWG

SEQ ID NO: 12: SADAEEDDSDVWWGG

SEQ ID NO: 13: EDDSDVWWGGADTDY

SEQ ID NO: 15: Ac-TETEFIAWGPTGDED-NH 2

SEQ ID NO: 16: Ac-EDDDWIDWYDSDDD-NH 2

SEQ ID NO: 17: Ac-EDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 18: Ac-EDDYIDWYDSDDD-NH 2

SEQ ID NO: 19: Ac-EDDYIDWGPTSDDD-NH 2

SEQ ID NO: 20: Ac-EDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 21 : Ac-EDDD(2Nala)IDWGPTSDDD-NH 2

SEQ ID NO: 22: Ac-EDDYTDWYDSDDD-NH 2

SEQ ID NO: 23: Ac-EDDYTDWGPTSDDD-NH 2

SEQ ID NO: 24: Ac-ETEFIAWGPTADED-NH 2

SEQ ID NO: 25: Ac-ETEYIAWGPTGDED-NH 2

SEQ ID NO: 26: Ac-ETEFIDWGPTGDED-NH 2

SEQ ID NO: 27: Ac-EDDSWIWWGPTGDED-NH 2

SEQ ID NO: 28: Ac-EDDSWIWWYGADTD-NH 2

SEQ ID NO: 29: Ac-EDDSWIDWGPTGDED-NH 2

SEQ ID NO: 30: Ac-EDDSWIDWYGADTD-NH 2

SEQ ID NO: 31 : Ac-ETEFI(Cya)WGPTGDED-NH 2

SEQ ID NO: 32: Ac-EDDD(Oetyr)IDWYDSDDD-NH 2

SEQ ID NO: 33: Ac-EDDD(Obtyr)IDWYDSDDD-NH 2

SEQ ID NO: 34: Ac-EDDD(Oetyr)IDWGPTSDDD-NH 2

SEQ ID NO: 35: Ac-EDDD(Obtyr)IDWGPTSDDD-NH 2 SEQ ID NO: 36: Ac-RRRRRRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 37: Ac-RRRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 38: Ac-RRREDDDWIDWYDSDDD-NH 2

SEQ ID NO: 39: Ac-RRRRRRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 40: Ac-RRRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 41 : Ac-RRREDDDWIDWGPTSDDD-NH 2

SEQ ID NO: 42: Ac-RRRREDDD(2Nala)IDWYDSDDD-NH 2

SEQ ID NO: 43: Ac-RRRREDDD(2Nala)IDWGPTSDDD-NH 2

SEQ ID NO: 22: Ac-EDDYTDWYDSDDD-NH 2

SEQ ID NO: 23: Ac-EDDYTDWGPTSDDD-NH 2

SEQ ID NO: 27: Ac-EDDSWIWWGPTGDED-NH 2

SEQ ID NO: 28: Ac-EDDSWIWWYGADTD-NH 2

SEQ ID NO: 48: Ac-EDDSDVWWGPTGDED-NH 2 (Peptide X)

SEQ ID NO: 49: Ac-ETEFIAWGGADTD-NH 2 (Peptide Y)

SEQ ID NO: 50: Ac-ETEFIAWYGADTD-NH 2 (Peptide 37)

SEQ ID NO: 51 : Ac-ETEFIAWG(/V-Me-G)TGDED-NH 2 (Peptide 38)

Tat-16 AC-YGRKKRRQRRREDDDWIDWYDSDDD-NH 2

16-Tat AC-DDDWIDWYDSDDDYGRKKRRQRRR-NH 2

SEQ ID NO: 55: Ac-EEDDSDVWWGGADTDYAD-NH 2 (Peptide 36)

Table 17. Overview of synthesized cyclic peptides. Data are displayed as calculated Kj values (mean ± SEM, n > 3).

References

Heaney, C. F.; Kinney, J. W., Role of GABA(B) receptors in learning and memory and neurological disorders. Neurosci Biobehav Rev 2016, 63, 1-28.

Bowery, N. G.; Bettier, B.; Froestl, W.; Gallagher, J. P.; Marshall, F.; Raiteri, M.;

Bonner, T. I.; Enna, S. J., International Union of Pharmacology. XXXIII. Mammalian y- aminobutyric acids receptors: structure and function. Pharmacol. Rev. 2002, 54 (2), 247-264.

Wedin, G. P.; Hornfeldt, C. S.; Ylitalo, L. M., The clinical development of y- hydroxy butyrate (GHB). Curr. Drug Saf. 2006, 1 (1), 99-106.

Bay, T.; Eghorn, L. F.; Klein, A. B.; Wellendorph, P., GHB receptor targets in the CNS: Focus on high-affinity binding sites. Biochem Pharmacol 2014, 87 (2), 220-228.

Gassmann, M.; Bettier, B., Regulation of neuronal GABAB receptor functions by subunit composition. Nat. Rev. Neurosci. 2012, 13 (6), 380-394.

Fritzius, T.; Bettier, B., The organizing principle of GABAB receptor complexes: Physiological and pharmacological implications. Basic Clin. Pharmacol. Toxicol. 2019. Bettier, B.; Tiao, J. Y., Molecular diversity, trafficking and subcellular localization of GABAB receptors. Pharmacol. Ther. 2006, 110 (3), 533-543.

Bettier, B.; Kaupmann, K.; Mosbacher, J.; Gassmann, M., Molecular structure and physiological functions of GABAB receptors. Physiol. Rev. 2004, 84, 835-867.

Vigot, R.; Barbieri, S.; Brauner-Osborne, H.; Turecek, R.; Shigemoto, R.; Zhang, Y. P.; Lujan, R.; Jacobson, L. H.; Biermann, B.; Fritschy, J. M.; Vacher, C. M.; Muller, M.; Sansig, G.; Guetg, N.; Cryan, J. F.; Kaupmann, K.; Gassmann, M.; Oertner, T. G.; Bettier, B., Differential compartmentalization and distinct functions of GABA(B) receptor variants. Neuron 2006, 50 (4), 589-601.

Blein, S.; Ginham, R.; llhrin, D.; Smith, B. O.; Soares, D. C.; Veltel, S.; Mcllhinney,

R. A. J.; White, J. H.; Barlow, P. N., Structural analysis of the complement control protein (COP) modules of GABAB receptor 1a J. Biol. Chem. 2004, 279 (46), 48292- 48306.

Ulrich, D.; Bettier, B., GABAB receptors: synaptic functions and mechanisms of diversity. Curr. Opin. Neurobiol. 2007, 17 (3), 298-303.

Perez-Garci, E.; Gassmann, M.; Bettier, B.; Larkum, M. E., The GABABW isoform mediates long-lasting inhibition of dendritic Ca 2+ spikes in layer 5 somatosensory pyramidal neurons. Neuron 2006, 50 (4), 603-616.

Schwenk, J.; Perez-Garci, E.; Schneider, A.; Kollewe, A.; Gauthier-Kemper, A.; Fritzius, T.; Raveh, A.; Dinamarca, M. C.; Hanuschkin, A.; Bildl, W.; Klingauf, J.; Gassmann, M.; Schulte, U.; Bettier, B.; Fakler, B., Modular composition and dynamics of native GABAB receptors identified by high-resolution proteomics. Nat. Neurosci. 2016, 19 (2), 233-242.

Dinamarca, M. C.; Raveh, A.; Schneider, A.; Fritzius, T.; Fruh, S.; Rem, P. D.;

Stawarski, M.; Lalanne, T.; Turecek, R.; Choo, M.; Besseyrias, V.; Bildl, W.;

Bentrop, D.; Staufenbiel, M.; Gassmann, M.; Fakler, B.; Schwenk, J.; Bettier, B., Complex formation of APP with GABAB receptors links axonal trafficking to amyloidogenic processing. Nat. Commun. 2019, 10, 1331.

Rice, H. C.; De Malmazet, D.; Schreurs, A.; Frere, S.; Van Molle, I.; Volkov, A. N.; Creemers, E.; Vertkin, I.; Nys, J.; Ranaivoson, F. M.; Comoletti, D.; Savas, J. N.;

Remaut, H.; Balschun, D.; Wierda, K. D.; Slutsky, I.; Farrow, K.; De Strooper, B.; De Wit, J., Secreted amyloid-p precursor protein functions as a GABAsRIa ligand to modulate synaptic transmission. Science 2019, 363 (6423), eaao4827.

Fritzius, T.; Bettier, B., The organizing principle of GABAB receptor complexes: Physiological and pharmacological implications. Basic Clin. Pharmacol. Toxicol. 2019. Conklin, B. R.; Farfel, Z.; Lustig, K. D.; Julius, D.; Bourne, H. R., Substitution of three amino acids switches receptor specificity of Gq alpha to that of Gi alpha. Nature 1993, 363 (6426), 274-6.

Yoo, Y.; Jung, J.; Lee, Y. N.; Lee, Y.; Cho, H.; Na, E.; Hong, J.; Kim, E.; Lee, J.

S.; Lee, J. S.; Hong, C.; Park, S. Y.; Wie, J.; Miller, K.; Shur, N.; Clow, C.; Ebel, R. S.; DeBrosse, S. D.; Henderson, L. B.; Willaert, R.; Castaldi, C.; Tikhonova, I.; Bilguvar, K.; Mane, S.; Kim, K. J.; Hwang, Y. S.; Lee, S. G.; So, I.; Lim, B. C.; Choi, H. J.; Seong, J. Y.; Shin, Y. B.; Jung, H.; Chae, J. H.; Choi, M., GABBR2 mutations determine phenotype in rett syndrome and epileptic encephalopathy. Annals of Neurology 2017, 82 (3), 466-478.

Garbutt JC, Approval of Baclofen for Alcohol Use Disorders in France: A Perspective From the United States. Alcohol and Alcoholism. 2020, 55(1) 48.