WO2013017631A1 - Kisspeptide-pentasaccharide conjugates - Google Patents

Kisspeptide-pentasaccharide conjugates Download PDF

Info

Publication number
WO2013017631A1
WO2013017631A1 PCT/EP2012/065061 EP2012065061W WO2013017631A1 WO 2013017631 A1 WO2013017631 A1 WO 2013017631A1 EP 2012065061 W EP2012065061 W EP 2012065061W WO 2013017631 A1 WO2013017631 A1 WO 2013017631A1
Authority
WO
WIPO (PCT)
Prior art keywords
pentasaccharide
kisspeptide
peptide
equiv
formula
Prior art date
Application number
PCT/EP2012/065061
Other languages
French (fr)
Inventor
Marion BLOMENRÖHR
Martin De Kort
Miranda Maria Cornelia VAN DER LEE
Jeffry Abraham Jacobus WISSE
Original Assignee
Msd Oss B.V.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Msd Oss B.V. filed Critical Msd Oss B.V.
Priority to US14/236,694 priority Critical patent/US9127038B2/en
Priority to EP12743963.6A priority patent/EP2755690B1/en
Publication of WO2013017631A1 publication Critical patent/WO2013017631A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • C07K9/001Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence having less than 12 amino acids and not being part of a ring structure

Definitions

  • Angew. Chem. Int. Ed. 49, 6288-6308, 2010 An alternative method for the prolongation of the half life of a peptide or a protein consist in the conjugation to a synthetic sulfated oligosaccharide, in particular a pentasaccharide, which has affinity to antithrombin III (AT III) as disclosed in WO 2006/082184 (Akzo Nobel N.V.) and in WO 2008/029278 (Endotis Pharma).
  • Antithrombin III is a serine protease inhibitor which is present in blood.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Reproductive Health (AREA)
  • Epidemiology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Endocrinology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention relates to kisspeptide-pentasaccharide conjugates having the general formula (I) wherein Ζ1 is Tyr or D-Tyr; Z3 is Trp, Hyp, Phe or Lys(R2); Z5 is Thr, Aib or Ala; Z7 is Gly or azaGly; Z8 is Leu; or Z7 and Z8 together represent; Z10 is Phe or Trp; n is 0 or 1; or R2, when present, represents a pentasaccharide derivative having the formula (II) wherein R is methyl or SO3X; X is a positively charged counterion; with the proviso that when R2 is present, R1 is H or (C1-6) alkylcarbonyl; R3 is H or (C1-3)alkyl; and L represents a pharmacologically inactive linker moiety having 10-50 atoms; or a pharmaceutically acceptable salt thereof; to pharmaceutical compositions comprising the same as well as to the use of said kisspeptide-pentasaccharide conjugates in the treatment of female infertility.

Description

KISSPEPTIDE-PENTASACCHARIDE CONJUGATES.
The present invention relates to kisspeptide-pentasaccharide conjugates, to pharmaceutical compositions containing said conjugates and to the use of the conjugates in the treatment of female infertility.
The most common cause of female infertility is ovulatory dysfunction. The most frequent being absent or irregular ovulation due to hypothalamic-pituitary dysfunction associated with normal basal levels of estradiol (WHO group II infertility) of which 80% of are known to be oligo/anovulatory due to polycystic ovarian syndrome (PCOS). The first line treatment for ovulation disorders comprises of a five day oral treatment regimen in the follicular phase of the menstrual cycle with the non-steroidal selective estrogen receptor modulator clomiphene citrate. Although ovulation rates are high after clomiphene citrate treatment (60%-90%), pregnancy rates are relatively low (20%-40%). The discrepancy is explained partly by prolonged anti-estrogenic effect of clomiphene citrate on endometrial receptivity and cervical mucus, due to the relatively long half-life of clomiphene citrate.
The KISS1-receptor (GPR54) plays a critical role in the central regulation of luteinizing hormone (LH) and follicle stimulating hormone (FSH) release (Roa et al, Endocrinology 147: 2864-2878, 2006). This has been validated by the infertile phenotype of KISS1 R mutations in human and targeted disruption in the KISS1 R gene in mice. On the other hand, activating mutations of KISS1 R cause precocious puberty. KISS1 R agonists lack anti-estrogenic activity and stimulate LH and FSH release, while the risk of overstimulation by kisspeptin is expected to be very low because the negative feedback at the pituitary level stays in tact. Therefore, KISS receptor agonists are perceived as potential new drugs for first line treatment of female infertility, such as ovulation induction (Ol), with improved implantation rate compared to clomiphene citrate, the current standard of care.
Kissl gene products such as Metastin (Kiss-54) and the Kisspeptins (kisspeptin-14, -13 and -10; for nomenclature see: Gottsch et al Peptides 30, 4-9, 2009) are potent endogenous peptide ligands of the KISS1-receptor, but their half-lives are considered too short for therapeutic applications. Kisspeptin-10 derivatives (also named metastin-derivatives or kiss-peptide derivative) having improved stability in blood were disclosed in WO 2006/001499 (Takeda Pharm. Co. Ltd). While the half-lifes of these derivatives in human plasma was enhanced from ~ 10 minutes for the native kisspeptin-10 to up to several hours, this stabilisation is not considered sufficient for effective applications in treatment of infertility because the small size of these stabilized analogs (< 2 kDa) does not prevent them from renal clearance, rendering them only effective after multiple repeated injections.
KISS1-peptide derivatives having enhanced half lifes resulting from covalent attachment of water soluble oligomers, such as polyethylene glycol (PEG), were disclosed in WO 2010/033224 (Nektar Therapeutics). Similarly, PEGylated metastin derivatives, such as kisspeptin-10 derivatives, having improved stability in blood were recently disclosed in WO 2010/013762 (Takeda Pharm. Co. Ltd.).
The in vivo use of PEGylated drugs, including proteins and peptides, is still associated with limitations such as the non-biodegradability of polyethylene glycol, the polydispersity of PEG, potential hypersensitivity reactions and unexpected change in pharmacokinetic behavior (Knop et al, " Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives";
Angew. Chem. Int. Ed. 49, 6288-6308, 2010). An alternative method for the prolongation of the half life of a peptide or a protein consist in the conjugation to a synthetic sulfated oligosaccharide, in particular a pentasaccharide, which has affinity to antithrombin III (AT III) as disclosed in WO 2006/082184 (Akzo Nobel N.V.) and in WO 2008/029278 (Endotis Pharma). Antithrombin III is a serine protease inhibitor which is present in blood. The serum half-life of the oligosaccharide-peptide conjugates was found to be largely determined by the half-life of the pentasaccharide, which is based on its affinity to ATMI. This directing effect of ATMI as a plasma protein carrier on the pharmacokinetic properties of oligosaccharide-peptide conjugates has been confirmed in studies of conjugates of pentasaccharides derived from the anticoagulant idraparinux with the 51-peptide insulin (de Kort et al. "Conjugation of ATIII-binding pentasaccharides to extend the half-life of proteins: long- acting insulin"; ChemMedChem 3, 1 189-1 193, 2008) and with the 36-peptide antiretroviral fusion inhibitor enfuvertide (Huet et al. "Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity", Antimicrob. Agents Chemother. 54, 134-142, 2010). There remains a need for kisspeptin derivatives having an enhanced in vivo half-life with retainment of potency and a predictable, safe and efficacious profile.
To that aim the present invention provides kisspeptide-pentasaccharide conjugates having the formula (I)
Ri -Zi -(Asn)n-Z3-Asn-Z5-Phe-Z7-Z8-Arg(R3)-Zi 0-NH2
Formula (I) wherein
Figure imgf000005_0001
Z3 is Trp, Hyp, Phe or Lys(R2);
Z5 is Thr, Aib or Ala;
Z7 is Gly or azaGly;
Z8 is Leu; or
Z7 and Z8 together represent
Figure imgf000005_0002
n is 0 or 1 ;
R-i or R2, when present, represents a pentasaccharide derivative having the formula (II)
Figure imgf000005_0003
Formula (II);
wherein R is methyl or S03X;
X is a positively charged counterion;
with the proviso that when R2 is present, R-i is H or (d-6) alkylcarbonyl;
R3 is H or (Ci-3)alkyl; and
L represents a pharmacologically inactive linker moiety having 10-50 atoms; or a
pharmaceutically acceptable salt thereof.
The kisspeptide-pentasaccharide conjugates of the invention display not only a vast prolonged half-life in human plasma, but are surprisingly found to retain their kisspeptin in vitro agonistic activity at the human KISS-1 receptor when measured in the presence of human serum (i.e. in the presence of ATMI) and to be extremely stabilized against metabolic degradation of the kisspeptide part of the conjugates of Formula (I). The kisspeptide-pentasaccharide conjugates of the invention have a therapeutic circulating plasma level of <50 nM. Up to this concentration the ATM I mediated anticoagulant activity of the oligosaccharide is insignificant. Thus, the oligosaccharide used in the conjugates of the present invention has an anticoagulant activity which is of subtherapeutic level when compared to the pharmacological activity of the kisspeptin component of the conjugate.
The term (d-3)alkyl as used in the definition of Formula (I) means a branched or unbranched alkyl group having 1 -3 carbon atoms, like propyl, isopropyl, ethyl and methyl.
The term (Ci_6)alkyl, as used in the term (Ci_6) alkylcarbonyl, likewise means a branched or unbranched alkyl group having 1-6 carbon atoms, like hexyl, pentyl, butyl, isobutyl, tertiary butyl, propyl, isopropyl, ethyl and methyl.
The pharmacologically inactive linker moiety having 10-50 atoms as used in the definition of Formula (II) means a flexible linking (or spacer) moiety which has 10-50 atoms counted along the "backbone" of the spacer.
The term "pharmacologically inactive" as used herein means that the linker does not contain atoms or groups which show pharmacologically activity per se at the doses at which the compounds of the invention are therapeutically effective. Thus, at doses at which the compounds of the present invention are used as therapeutic drugs, the nature of the spacer does not lead to demonstrable pharmacological side-effects.
The linker structure covalently links the pentasaccharide derivative in Formula (II) at the 4- position of the non-reducing end to an amine function of the kisspeptin-10 peptide analog. This amine function can be the a-amino group of the amino acid residue Z-i of Formula (I) or the ε- amino group of the compound of Formula (I) wherein Z3 represents a residue of lysine.
The linker moiety can comprise 10-50 atoms, more preferably 10-35 atoms and even more preferably 15-30 atoms.
Suitable linkers L comprise one or more (oligo)ethylene glycol groups such as exemplified by:
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-, -CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-CH2CH2CH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2SCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2SCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)mC(0)CH2CH2SCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NH-U-,
-(CH2CH20)nCH2CH2NH-U-NH-(CH2CH20)mCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)m-CH2CH2NH-U- wherein m, n or q may be 0-30;
T is 1 ,2,3-triazol-1 ,4-diyl;
U is cyclobut-3-ene-1 ,2-dione-3,4-diyl; and wherein the left side of the linker as depicted is attached to the pentasaccharide part of Formula (II) and its right side is attached to the Kisspeptide part of Formula (I).
Preferred are linkers L selected from:
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-, -CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-CH2CH2CH2CH2C(0)-,
wherein m, n or q may be 0-30; and
T is 1 ,2,3-triazoM ,4-diyl.
More preferred linkers are:
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-, -CH2CH20)nCH2CH2NHC(0)CH20(CH2CH20)qC(0)CH2CH2-T-(CH2CH20)mCH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-CH2CH2CH2CH2C(0)-;
wherein n= 2-10, m =2-10 and q= 2-10; and
T is 1 ,2,3-triazoM ,4-diyl.
Particular preferred linkers are
- (CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2CH2C(0)-,
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-(CH2CH20)mCH2C(0)- and
-(CH2CH20)nCH2CH2NHC(0)CH2CH2-T-CH2CH2CH2CH2C(0)-;
wherein n=2-4 and m= 0-4; and
T is 1 ,2,3-triazoM ,4-diyl.
There is a preference for kisspeptide-pentasaccharide conjugates of Formula (I), wherein Z5 is Thr. Further preferred are the kisspeptide-pentasaccharide conjugates of Formula (I), wherein in addition R3 is methyl. Also preferred are the conjugates of Formula (I) wherein also n is 0; Z-i is D- Tyr; Z3 is Hyp and Z10 is Trp. Other preferred conjugates of the invention are those of Formula (I) wherein n is 1 and Z 0 is Phe. Further preferred are the kisspeptide-pentasaccharide conjugates of Formula (I), wherein R is methyl, corresponding to conjugates wherein the pentasaccharide derivative contains 6 sulfate groups. Conjugates of the invention wherein R is methyl generally have a diminished affinity of the conjugate for ATIII and a resulting enhancement in potency in the presence of serum, as compared with the conjugates of Formula (I) wherein R is S03X. Specifically preferred kisspeptide-pentasaccharide conjugates of the invention are:
Conjugate C7
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
 Conjugate C41
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof.
The kisspeptide-pentasaccharide conjugates of the invention may be prepared by methods known in the art of bio-organic chemistry in general.
The conjugates of the invention according to Formula (I) can be prepared by a stepwise assembly of an optionally modified kisspeptide (KP) analog and a synthetic pentasaccharide (PS) derivative, followed by a process in which the linker L is formed by a coupling step linking the kisspeptide analog and the pentasaccharide derivative by a covalent bond.
The synthetic pentasaccharide (PS) derivative can be represented by Formula (III), wherein R represents methyl or S03X; X is a positively charged counterion;
Figure imgf000011_0002
OMe OMe
Formula
M represents part of the pharmacologically inactive linker moiety L, where L has the meaning as earlier defined; and P is a functional group, such as NH2, N3, thiol, alkyn, bromoacetyl and the like, which can engage in a conjugation reaction with a modified kisspeptide (KP) analog of either Formula (IV) or Formula (V), wherein Z-i , Z3, Z5, Z7, Z8, Z10, R3, n and M have the earlier defined meanings and wherein P-i and P2 independently represents a functional group, such as NH2, N3, thiol, alkyne, bromoacetyl and the like.
P1-M-Z1-(Asn)n-Z3-Asn-Z5-Phe-Z7-Z8-Arg(R3)-Z1o-NH2
(Formula IV);
Ri-Z1-(Asn)n-Lys(M-P2)-Asn-Z5-Phe-Z7-Z8-Arg(R3)-Z1o-NH2
(Formula V);
The conjugation method may comprise (a) an optional step wherein the kisspeptide analog is adapted for conjugation with a reactive group, (b) an optional step wherein the pentasaccharide derivative is adapted for conjugation with a reactive group and either (c) a coupling step wherein the optionally adapted kisspeptide analog is conjugated with the pentasaccharide-spacer molecule, wherein the pentasaccharide is extended at the non-reducing end with a spacer that is optionally modified with a reactive group, or (d) a step in which the kisspeptide analog and the pentasaccharide derivative are linked by a coupling reagent.
Optional modifications of and processes for linking the kisspeptide analog and the pentasaccharide derivative can be accomplished by well known methods in the art. General synthetic methods for the production of bioconjugates are described in "Bioconjugate Techniques" by Greg T. Hermanson, 2nd edition, 2008, Academic Press and "Bioconjugation Protocols" by Christof M. Niemeyer, 2004, Methods in Molecular Biology, Vol. 283, Humana Press; "Chemical Reagents for Protein Modification" by Roger L. Lundblad, 3rd edition, 2005, CRC Press; D. Crich, Reagents for Glycoside, Nucleotide and Peptide Synthesis, Wiley, Chichester, 2005 and M.A. Gauthier et al. Peptide/protein-polymer conjugates: synthetic strategies and design concepts Chem. Commun. 2008, 2591-261 1 . In addition, a Staudinger ligation (such as described by K.L. Kiick et al. Proc. Nat. Acad. Sci. 2002; 99:19-24) may be considered for conjugation. Alternatively, enzymatic reactions such as the regioselective IgA protease mediated elongation of polypeptides at the N-terminus (as described by M. Lewinska et al. in Bioconjugate Chem. 2004; 15: 231-234) or the transglutaminase catalyzed introduction of amino spacer containing oligosaccharides (as described by M. Sato et al. in J. Am. Chem. Soc. 2004; 126: 14013-14022) can be adapted for conjugation of a pentasaccharide spacer residue to an optionally modified kisspeptide analog.
In one method of conjugation a pentasaccharide derivative having a (protected) thiol function is coupled to a kisspeptide analog that contains a halide function, such as a bromoacetyl group as described in P. Schelte et al. Bioconjugate Chem. 2000, 1±, 1 18-123. Thiol-bromide conjugation method (A)
PS-(CH2CH20)nCH2CH2NHC(0)CH2CH2S(CO)CH3 + BrCH2C(0)-KP
J NH2OH
PS-(CH2CH20)nCH2CH2NHC(0)CH2CH2-S-CH2C(0)-KP thio-ether linkage
wherein PS represents the pentasaccharide part of Formula (II) and KP represents the Kisspeptide part of Formula (I).
In an alternative method of conjugation, a Huisgen's 1 ,3-dipolar cycloaddition to form a triazole linker is used for conjugation of a pentasaccharide derivative and a kisspeptide analog, that have been independently modified with an alkyne or azide functional group. The 1 ,3-dipolar
cycloaddition can be accomplished in a copper(l) catalyzed fashion and can optionally be accelerated by the use of a tailored ligand such as described in S.I. Presolski et al. J. Am. Chem. Soc. 2010, 132, 14570-14576; and by Jean-Francois Lutz, " 1,3-Dipolar Cycloadditions of Azides and Alkynes A Universal Ligation Tool in Polymer and Materials"; Angew. Chem. Int. Ed. 2007, 46, 1018 - 1025.
Cycloaddition conjugation method (C)
PS-(CH2CH20)nCH2CH2NHC(0)CH2CH2CCH N3-(CH2CH20)mCH2CH2C(0)-KP alkyne azide
I
N=N
PS-(CH2CH20)nCH2CH2NHC(0)CH2CH2 ^^ — (CH2CH20)mCH2CH2C(0)-KP
1 ,2,3-triazol-1 ,4-diyl linkage
Alternatively, metal-free methods of triazole linker formation can be applied as described in M.F. Debets et al. ChemBioChem 2010, 1±, 1 168-1 184.
In another method of conjugation, a kisspeptide (KP) analog having a primary amine function and a synthetic pentasaccharide (PS) derivative having a primary amino function can be conjugated with the use of a homobifunctional reagent, such as for example 1 ,2- diethoxycyclobutene-3,4-dione (squaric acid diester; Lutz et al., Bioconjugate Chem. 2, 148-153, 1991 ). Squarate conjugation method (B)
PS-(CH2CH20)nCH2CH2NH2 H2NCH2CH2C(0)-KP
Figure imgf000014_0001
squarate linkage
Further alternative bioorthogonal conjugation reactions were recently reviewed by Michael D. Best, "Click Chemistry and Bioorthogonal Reactions: Unprecedented Selectivity
in the Labeling of Biological Molecules", Biochemistry 2009, 48, 6571-6584. The required kisspeptide (KP) analog of either Formula (IV) or Formula (V) can be prepared by methods known in the art, such as solution phase peptide synthesis and manual or automated solid phase peptide synthesis (SPPS), or by a combination thereof.
The peptide coupling can be carried out by methods commonly known in the art for the coupling - or condensation - of peptide fragments such as by the azide method, mixed anhydride method, activated ester method, the carbodiimide method using DCC, or carbodiimide hydrochloride (EDCI.HCI); or, preferably, under the influence of ammonium/uronium salts like 2-(1 H- benzotriazol-1-yl)-1 , 1 ,3,3-tetramethyluronium tetraflluoroborate (TBTU), especially with the addition of catalytic and racemisation suppressing compounds like N-hydroxy-succinimide, N- hydroxybenzotriazole and 7-aza-N-hydroxybenzotriazole (HOBt, HOAt, HOOBt, etc.). Overviews are given in The Peptides, Analysis, Synthesis, Biology, Vol. 3, E. Gross and J. Meienhofer, eds. (Academic Press, New York, 1981 ), Peptides: Chemistry and Biology, N. Sewald and H.-D. Jakubke (Wiley-VCH, Weinheim, 2002) and Chemistry of Peptide Synthesis, N.L.Benoiton (CRC Taylor & Francis, 2005).
SPPS is well known in the art (Merrifield, R. B. J. Am. Chem. Soc, 85, 2149-2154 (1963), and is widely employed by commercial suppliers. (See also Sewald, N. Peptides: Chemistry and Biology, Wiley-VCH, 2003 and Bodanszky, Principles of Peptide Synthesis, Springer-Verlag, Heidelberg (1984)). There are several known variations on the general approach. (See, for example, "Peptide Synthesis, Structures, and Applications" 1995 by Academic Press, Chapter 3 and White (2003) Fmoc Solid Phase Peptide Synthesis, A practical Approach, Oxford University Press, Oxford).
Methods for obtaining a peptide in the form of a C-terminal amide, such as in peptide derivatives of Formula (I), include, for example, solid phase synthesis using resins (e.g Rink amide) for the formation of peptide amides. Alternatively, the carboxyl group of the terminal amino acid is first protected by amidation and the peptide chain is subsequently extended from the amino group side to a desired length.
In an alternative fashion, the peptide can be assembled by classical solution phase synthesis or in a combination of classical SPPS and solution phase synthesis, such as as described in I.F. Eggen et al. J. Peptide Sci. 11 , 633-641 (2005), I.F. Eggen et al. Org. Proc. Res. Dev., 9, 98-101 (2005) and EP1277761.
After completion of the reaction on the solid support or in solution, the product may be purified and isolated by a combination of conventional purification methods such as solvent extraction, column chromatography, liquid chromatography, trituration and recrystallization. When the peptide obtained by the above methods is in a free form, the peptide can be converted into an appropriate salt; conversely when the peptide is obtained in a salt form, it can be converted into its free form or an alternative salt form by methods known in the art.
When the kisspeptide analog of the conjugate of the invention is present in the form of a configu rational isomer, each diastereomer or conformer can optionally be isolated by separation and purification as mentioned above. In addition, when the compound of the present invention is racemic, it can be separated into its enantiomers by the conventional means of optical resolving.
The kisspeptide analog may optionally contain stabilizing and/or potency enhancing structural elements or the peptide isosteric replacements such as aza-glycine or triazole moieties. The preparation of azapeptides by SPPS or solution phase synthesis is well known in the art. For introduction of aza-glycine, 4-nitrophenyl chloroformiate and Fmoc-NH-NH2.TFA can be used to extend the optionally immobilized tripeptide intermediate at the N-terminus as in Formula VI. a) 4-N02PhOCOCI + H-Leu-Arg(R3)-Z10-RinkResin
- 4-N02PhOC(0)-Leu- Arg(R3)-Z10-RinkResin b) 4-N02PhOC(0)-Leu- Arg(R3)-Z10-RinkResin + Fmoc-NH-NH2.TFA
Fmoc-NH-NH-CONH-L-R-F-RinkResin
Formula (VI) Alternatively, the reagents in Formula (VII) can be prepared for introduction of the aza-glycine using phosgene according to J. Org. Chem. 64, 7388 (1999).
Figure imgf000016_0001
Formula (VII) In addition, the aza-kisspeptide can be assembled in a chemoselective silver catalyzed coupling of a thioester derivative according to N. Olivier et al. Bioconjugate Chem. 2009, 20, 1397-1403. Subsequently, the Fmoc-protected aza-Gly intermediate can be converted into a nonapeptide or decapapetide kisspeptide analog by means of known methods for solution or solid phase peptide synthesis. Suitable aza-kisspeptide analogs for compounds of the present invention have been described in WO2004063221 or WO2006001499.
Peptide isosteric elements, such as the 1 ,4-(1 ,2,3-triazole) group in the definition of Formula (I) wherein Z7 and Z8 together represent
Figure imgf000016_0002
can be introduced by the condensations of azide and alkyne containing peptide fragments as in A. Brik et al. Chem. BioChem 6, 1 167 -1 169 (2005) or J. Springer, et al. Eur. J. Org. Chem. 15, 2592-2600 (2008). Preferably, the azide is present at the N-terminal position of the growing peptide chain and the alkyne is present at the C-terminal end of the incoming peptide building block or peptide chain as in Formula (VII), wherein R5 may be a suitable temporary amino protecting group (such as Fmoc or Boc) and R6 is H, a suitably amide protecting group or a covalent linkage to a resin from which the kisspeptide analog is cleaved according to a process as described above. Similarly, the introduction and of the azide function and the subsequent cycloaddition can be accomplished as above. The growing triazole kisspeptide analog can then be further processed either by solution phase or solid phase peptide synthesis, or a combination thereof, into the peptide analogs as depicted in Formula (IV) and (V). AND Enantiomer
Figure imgf000017_0001
Formula (VIII)
The ATIII-binding pentasaccharide derivative according to Formula (III) can be prepared using methods described by Van Boeckel and Petitou in Angew. Chem. Intl. Ed. Engl. 32, 1671-1690 (1993) and by C. Chen et al. in Biorg. Med. Chem. Lett. 19, 3875-3879, 2009.
The kisspeptide-pentasaccharide conjugates of the present invention may be purified according to well-known methods as described in H. Ahmed, Principles and Reactions of Protein Extraction, Purification and Characterization, CRC Press, 2005, M.-l. Aguilar, HPLC of Peptides and Proteins, Methods and Protocols, Humana Press, New Jersey, 2004 and K.M. Gooding and F.E. Regnier, HPLC of Biological Macromolecules, Marcel Dekker Inc., New York, 2002. Preferred methods of purification involve anion and/or cation exchange chromatography, hydrophobic interaction chromatography, size exclusion chromatography and high performance liquid chromatography (HPLC). Most preferred methods of purification include anion exchange chromatography and HPLC.
Furthermore, the compounds of the invention, which may occur in the form of a free base, may be isolated from the reaction mixture in the form of a pharmaceutically acceptable salt. The pharmaceutically acceptable salts may also be obtained by treating the free base of Formula (I) with an organic or inorganic acid such as hydrogen chloride, hydrogen bromide, hydrogen iodide, sulfuric acid, phosphoric acid, acetic acid, propionic acid, glycolic acid, maleic acid, malonic acid, methanesulphonic acid, fumaric acid, succinic acid, tartaric acid, citric acid, benzoic acid, ascorbic acid and the like.
The kisspeptide-pentasaccharide conjugates of this invention or intermediates thereof may possess chiral carbon atoms, and may therefore be obtained as a pure enantiomer, or as a mixture of enantiomers, or as a mixture containing diastereomers. Methods for obtaining the pure enantiomers are well known in the art, e.g. crystallization of salts which are obtained from optically active acids and the racemic mixture, or chromatography using chiral columns. For diastereomers straight phase or reversed phase columns may be used. The present invention further provides pharmaceutical compositions comprising a kisspeptide- pentasaccharide conjugates according to Formula (I), or a pharmaceutically acceptable salt thereof, in admixture with pharmaceutically acceptable auxiliaries, and optionally other therapeutic agents. The term "acceptable" means being compatible with the other ingredients of the composition and not deleterious to the recipients thereof. Compositions include e.g. those suitable for parenteral administration, such as intravenous, intraperitonial, subcutaneous, intramuscular, transdermal administration, and the like. Preferred routes of administration are the subcutaneous and intramuscular route.
Pharmaceutical compositions for parenteral administration may contain 0.01-25 mg/ml of a kisspeptide-pentasaccharide conjugate in an aqueous solution buffered at a pH between 3 and 8, preferably between 4-6. A preferred composition of the invention comprises an aqueous buffered solution comprising 5 % (w/w/) mannitol and 6 mM sodiumacetate buffered at pH 5. For parenteral administration, the pharmaceutical composition of the invention may be presented in unit-dose or multi-dose containers, e.g. injection liquids in predetermined amounts, for example in sealed vials and ampoules, and may also be stored in a freeze dried (lyophilized) condition requiring only the addition of sterile liquid carrier, e.g. water, prior to use.
For parenteral administration, aqueous suspensions, isotonic saline solutions and sterile injectable solutions may be used, containing pharmaceutically acceptable dispersing agents and/or wetting agents, such as propylene glycol or butylene glycol, as described in the standard reference, Gennaro, A.R. et al, Remington: The Science and Practice of Pharmacy (20th Edition, Lippincott Williams & Wilkins, 2000, see especially Part 5: Pharmaceutical Manufacturing), The invention further includes a pharmaceutical composition, as hereinbefore described, in combination with packaging material suitable for said composition, said packaging material including instructions for the use of the composition for the use as hereinbefore described.
The conjugates of the invention may be administered to humans in a unit dose comprising 0.01- 10 mg of conjugate, preferably 0.1-5 mg and more preferred 0.1-2 mg. In a preferred regimen for the treatment of female infertility a single unit dose per menstrual cycle is applied
subcutaneously.
The kisspeptide-pentasaccharide conjugates of the invention were found to be agonists of the KISS receptor.
In vitro assays to determine the biological activity of KISS receptor agonists are well-known in the art. In general, cells expressing the KISS receptor are incubated with the compound to be tested and the stimulation of a functional response is determined. To measure a functional response, isolated DNA encoding the KISS receptor gene, preferably the human receptor, is expressed in a suitable host cell-line. Such a host cell-line might be the Chinese Hamster Ovary (CHO) cell-line, but other cell-lines can also be used. Preferably, the host cells are of mammalian origin (Jia et al (1991 ) Mol Endocrinol 5, 759-776).
Methods to construct KISS receptor-expressing cell lines are well-known in the art (e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, latest edition). Heterologous expression of the receptor is obtained by transfection and expression of the DNA encoding the desired protein. Techniques for PCR and construction of suitable expression systems are also well-known in the art. The DNA encoding the desired protein can be constructed synthetically using standard solid phase techniques, preferably to include restriction sites for ease of ligation. Suitable control elements for transcription and translation of the included coding sequence can be provided to the DNA coding sequences. As is well-known, expression systems are available, which are compatible with a wide variety of hosts, including prokaryotic hosts such as bacteria and eukaryotic hosts such as yeast, plant cells, insect cells, avian cells, mammalian cells, and the like.
Cells expressing the receptor are then incubated with the test compound to determine stimulation of a functional response.
The KISS receptor is a family-A G-protein coupled receptor (GPCR) that couples via the Gaq- protein to the Phospholipase C/Calcium2+ signal transduction pathway. The generation of Calcium2+ in the presence of a KISS agonists can be determined in the FLIPR assay, in which a Calcium2+ sensitive fluorescent dye is used as a sensor for intracellular Calcium2+ (Hansen KB and Brauner-Osborne H (2009) Methods Mol Biol 552, 269-78). Upstream of Calcium2+, the Inositol Phosphate cascade can be measured by detection of IP1 accumulation in cells using Homogeneous Time-Resolved Fluorescence (HTRF) assay (Cassutt KJ et al (2007) J of Biomol Screening 12, 285-287).
In addition to the direct measurement of Calcium2+ levels in the KISS receptor-expressing cell- line, cell-lines may be transfected with a second cDNA that encodes a reporter gene, of which the expression is dependent on the intracellular concentration of Calcium2+. In general, reporter gene expression might be controlled by any response element reacting to changing levels of intracellular Calcium2+ e.g. NFAT-response element. Suitable reporter genes are e.g. the genes encoding beta-galactosidase, alkaline phosphatase, firefly luciferase and green fluorescence protein. The principles of such transactivation assays are well-known in the art and are described for example in Stratowa et al (1995) Curr Opin Biotechnol 6, 574. Agonistic compounds may also be identified in assays that are based on receptor-induced recruitment of beta-arrestin to the agonist-occupied receptor (e.g., Transfluor® assay, PathHunter® and Tango™ beta-arrestin assays) or receptor internalization assays (e.g., PathHunter® endocytosis assays). Label-free assays may also be applicable to screen for KISS receptor agonists. These assays are based on receptor-induced dynamic mass redistribution of intracellular content or receptor-induced changes in cell morphology or adhesion (Van Koppen (2010) "Generic Assays in G-protein- coupled Receptor Drug Discovery"; Drug Discovery 7, 69-72).
The kisspeptide-pentasaccharide conjugates of the invention showed in vitro agonistic activity at the Gaq-coupled human KISS receptor, as measured in the absence and in the presence of 20% human serum in a NFAT-regulated luciferase reporter gene assay. When tested at 10~8 M in the presence of 20% human serum the conjugates of the invention demonstrates more than 50% stimulation of the effect induced by Kisspeptin-10. A pEC50 of higher than 8 in the presence of 20% human serum (in the presence of ATIII) indicates that the conjugates of the invention may be active in vivo at a human dose devoid of anti-coagulant activity.
The kisspeptide-pentasaccharide conjugates of the invention were found to be remarkably stable against proteolytic degradation upon in vitro stability testing in human serum (Tomita et al, (2008) J. Med. Chem. 51 (23), 7645-7649). Moreover the kisspeptide-pentasaccharide conjugates were also found to be stable in vivo as determined in pharmacokinetic studies.
Assays to detect levels of intact kisspeptide-pentasaccharide conjugates in human serum (both from in vitro serum stability testing and in vivo PK studies) are well-known in the art, such as assays method employing an antibody directed against the conjugate (e.g. in an Enzyme-linked immunosorbent assay, ELISA or Radio Immuno Assay, RIA, Dhillo W et al (2007) J Clin
Endocrinol Metab 92, 3958-3966), in a competition assay with radioactive-labeled Kisspeptides (e.g. a KISS receptor binding assay or RIA), in a bio-activity assay (e.g. Calcium2+ measurements in cells expressing the KISS receptor, see above) or by liquid chromatography-mass
spectrometry (LC-MS, McCormack AL et al. (1997) Anal. Chem. 69 (4), 767-776).
Conjugates of the present invention are characterized by a potency that is sufficient to provide effective therapeutic plasma levels <50 nM and an in vitro serum stability of the conjugate that matches the in vivo half-life (> 24-120h in human). The kisspeptide-pentasaccharide conjugates of the invention can be used in the treatment of female infertility. The conjugates can have further utility in the treatment of (metastatic) cancer (especially hormone-dependent cancers like prostate cancer and breast cancer) and in the treatment of endometriosis and hot flushes in breast cancer. In addition, the kisspeptide- pentasaccharide conjugates of the invention can also be used in other mammals than human. Experimental
ABBREVIATIONS
Aib aminoisobutyric acid
ACN acetonitrile
Boc t-butyloxycarbonyl
BSA bovine serum albumin
CHO Chinese hamster ovary
CV column volume
DCM dichloromethane
DIEA diisopropyl ethyl amine
DIC diisopropyl carbodiimide
DMF N,N-dimethylformamide
EC50 effective concentration resulting in 50% of the maximum response
EDCI 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
ESI electrospray ionization
Fmoc 9-fluorenylmethoxycarbonyl
HPLC high pressure liquid chromatography
HOBt 1 -hyd roxy be nzotriazole
Hyp 3-hydroxyproline
(LC)MS (liquid chromatography) mass spectrometry
Mbs p-methoxybenzenesulfonyl
MTBE tert-butyl methyl ether
Mtr 4-methoxy-2,3,6-trimethylbenzenesulfonyl
Mts mesitylene-2-sulfonyl
NMM N-methyl morpholine
Np para-n itrophenol
Pbf 2,2,4,6,7-pentamethyldihydrobenzofuran-5-sulfonyl
Plmc 2,2,5,7,8-pentamethylchroman-6-sulfonyl
RP reversed phase
Sc subcutaneous
TBTU 2-(1 H-benzotriazol-1-yl)-1 ,1 ,3,3-tetramethyluronium tetrafluoroborate
TFA trifluoroacetic acid
Tos tosyl
Trt trityl
UV ultraviolet
Vis visible
Z benzyloxycarbonyl MATERIALS AND METHODS
Analytical methods
HPLC analysis
Table : LC methodology for purity determination (representative method A)
Figure imgf000022_0001
Method B: A: 25 mM NH4Ac in H20 + 10% ACN, B: ACN + 10% 25 mM NH4Ac in H20. 0->80%B in 30min, flow 0.25 mL/min)
Method C: A: 25 mM NH4Ac in H20 + 10%ACN, B: ACN + 10% 25 mM NH4Ac in H20. 10- >50%B in 30min, flow 0.25 mL/min
Method D: A: 25 mM NH+Ac in H20 + 10% ACN, B: 25 mM NH4Ac in ACN/H20 8/2. 10->50%B in 30min, flow 0.25 mL/min
Method E (applied with LCMS): A: 25 mM NH4Ac in H20/ACN 9:1 v/v, B: 25 mM NH^c in H20/ACN 1 :9 v/v. 10 -> 80%B in 30 min Mass spectrometry analysis
Absorption spectra were recorded on a ATI Unicam Ltd. UV2 UV/Vis spectrometer. The spectrometer was controlled via Vision 3.4 software running on a personal computer. Typical experimental parameters were 2 nm resolution and using a computerized optimal scan time. High-resolution MS spectra were acquired on a Bruker Daltonics MicroTOF-Q time-of-flight mass spectrometer. Spectra were recorded using electrospray ionization (ESI) in positive ion mode unless indicated otherwise. The instrument was controlled via HyStar 3.2 software running on a personal computer. Typical experimental parameters for MS were: 200°C Dry Heater temperature. Nitrogen gas was used as both drying and nebulising gas, where the drying gas flow rate was 4.0 L«min-1 and the nebuliser pressure 1.4 bar. For positive ion mode, summation of 5000 spectra were acquired with a window of 300-3000 m/z. External calibration was obtained by infusing a mixture of Agilent tune mix 1 : 100 in ACN for 0.5 min at the end of the experiment. The nominal resolution of the instrument was 19000, FWHH, the accuracy <5 ppm. A Dionex Ultimate 3000 HPLC system was used for injection at a flow rate of 50 ul«min-1 ; for chromatographic method descriptions see below. Typical sample concentrations were 0.5 mg/ml.
Synthetic Methods
Figure imgf000024_0001
8
a: ho^O^0^O^0^N3 b: c: ΗΟΓ -°-0-°-8ΑΟ d: HO^
Scheme I. Preparation of pentasaccharide derivatives 1 -10.
Pentasaccharides 1 , 2 and 5 were prepared as described in International Patent Application WO2006/082184 (Akzo Nobel N.V.) Pentasaccharide 3
14-azido-3,6,9, 12-tetraoxatetradecan-1-oic acid (compound a, 0.581 mmol, 161 mg) was dissolved in 8 mL DMF and NMM (0.953 mmol, 0.105 ml, 96 mg) was added, followed by the addition of TBTU (0.572 mmol, 184 mg). The mixture was stirred for 20 min. Then Pentasaccharide 1 (0.318 mmol, 600 mg) was added as a solid. The mixture was stirred for another 2 h and was subsequently concentrated under reduced pressure. The residue was dissolved in water, filtered over PVDF 0.45 urn filter and purified by Q-Sepharose® ion exchange chromatography. The fractions containing conjugated azide were collected and desalted by Sephadex® G-25 gel filtration chromatography. Yield: 650 mg (95%). Next, the conjugated azide (0.088 mmol, 190 mg) was dissolved in t-BuOH/water (1 :1 , v/v, 20 mL). The solution was degassed, placed under an atmosphere of nitrogen and Pd/C (0.422 mmol, 50 mg) was added. The heterogeneous mixture was degassed and placed under an atmosphere of hydrogen which was lead through the solution
for 18 h. The hydrogen gas was replaced by nitrogen and the solution was filtered and concentrated under reduced pressure to give Pentasaccharide 3 as the free amine in 90% yield. Using the same procedure pentasaccharide 4 was prepared staring from pentasaccharide 2.
Pentasaccharide 6
Pentynoic acid (compound b; 1.455 g, 14.84 mmol, 1.5 eq) was dissolved in DMF (15 mL). HOBt (2.205 g, 16.32 mmol, 1.65 eq) and EDCI (2.84 g, 14.84 mmol, 1.5 eq) were added and stirred for 1 h at r.t. A solution of Pentasaccharide 1 (18.68 g, 9.89 mmol, 1 eq) in DMF (200mL) was added drop wise the reaction mixture and stirred for 2 h. Water (300 mL) was added before the reaction mixture was extracted with DCM 3 times. The water layer was filtered and the remaining DCM was removed under reduced pressure. The remaining water layer was eluted on to a Q-Sepharose column (CV 450 mL) and eluted with 0.2 M NaCI(aq) (2 CV), 0.2 M to 1.0 M (20 CV) and 2.0 M (2 CV). The solution collected was concentrated under reduced pressure to a smaller volume before desalting on a Sephadex G25 column. After lyophilizing the solution, compound 6 (16.5 g, 8.38 mmol 85%) was isolated.
Using the same procedure pentasccharide 7 was prepared staring from pentasccharide 2.
Pentasccharide 9 or 10 were prepared from pentasccharides 3 or 4, respectively, as described for the conversion of the pentasccharide 1 or 2 into 6 or 7, respectively. Synthesis of kisspeptides:
Kisspeptide analogues of Formula (IV) and Formula (V) were prepared by using solid phase peptide synthesis by commercial suppliers. Kisspeptides analogues wherein Z7= azaGly were prepared as described in WO2007072997, WO2006001499 or WO2004063221.
Kisspeptide analogues of Formula (IV) or Formula (V) wherein Z7 and Z8 together represent
Figure imgf000026_0001
, were prepared by incorporation of azidoleucine according to Org. Lett. 2001 , 3, 781-783 or by conversion of leucine into azidoleucine according to Org. Lett. 2007, 9, 3797-3800, followed by formation of the triazole peptide according to J. Org. Chem. 2007, 72, 7963-7967 or Org. Lett. 2006, 8, 4505-4507. Kisspeptides analogues wherein R3=Me were prepared by incorporation of Fmoc-Arg(Me,Pbf)-OH as a commercially available amino acid building block. Kisspeptides analogues wherein Z3= azidolysine were prepared by incorporation of commercially available amino acid building block Fmoc-azido-lysine.
Introduction of the P-i-M group as defined in Formula (IV) was performed either as the penultimate step in the solid phase synthesis procedure by an acylation step with the appropriate commercially available linking reagent (e.g. azidopentanoic acid), or by subjecting the peptide in solution to the corresponding mixed anhydride derivative that was freshly prepared as described for instance in Peptides Volume 5, Issue 2, 1984, 167-168.
Table A
Kisspeptides analogues: Pi-M-Z1-(Asn)n-Z3-Asn-Z5-Phe-Z7-Z8-Arg(R3)-Z1o-NH2
Formula (IV)
Figure imgf000027_0001
Figure imgf000028_0001
Representative example P4:
Modified kisspeptide analogue P4 was manufactured by solid phase peptide synthesis (SPPS) using the Fmoc (9-fluorenylmethyloxycarbonyl) strategy. All the deprotections were made with piperidine 35% in DMF. The C-terminal phenyl residue is coupled to the resin (p- methylbenzhydrylamine polystyrene-1 % divinylbenzene) via a Rink amide linker. The other amino acid residues are incorporated by a succession of Fmoc deprotection and amino acid coupling cycles. The summary of coupling amino acids on a resin (coupling method, excess and the time of coupling) is described in table B below:
Table B
Figure imgf000028_0002
After solid phase assembly, cleavage from the resin and concomitant side chain deprotection with a mixture of TFA/H20/ thioanisole (90/5/5) during 2h30 hours at room temperature yields directly to the crude peptide with the C-terminal amide. After concentration of -1/3 of the volume of the crude product mixture, the crude product is precipitated in cold MTBE. The precipitated peptide is filtered, washed with MTBE and dried under vacuum. The use of TFA/H20 /thioanisole gave the best results to reduce of the formation of peptide-Rink Linker and suppresses the conversion of the azide into an amino group. Purification is performed by preparative reverse phase HPLC in TFA buffer. The eluting fractions are analyzed. The pure fractions are pooled daily before lyophilisation. Each lot of purified peptide thus obtained is tested by HPLC against the specification before entering the final lot. The selected pools are mixed together to form a homogeneous solution in water before freeze drying. Optional ion exchange into an acetate salt is performed on a Dowex resin after which the final solution is freeze dried and packaged.
Purification is performed by a preparative reverse phase HPLC. 1 g of crude peptide was injected per run (dissolved in 100 mL of AcOH /H20/ACN 10/60/30). After filtration this mixture was loaded on the column. Buffer A= 0.1 % of TFA in water. Buffer B= ACN/buffer A (60/40 v/v). Stationary phase: C18 120A 15μιη. Column: 200 x 50mm. Flow rate: 100 mL/min. UV detection: 210 nm. Linear gradient from 40 to 65% B in 30 min. The fractions are classified into waste, semi pure or pure fractions (waste: < 70%, pure: >97%, front and back impure: 70% to < 97%). Front and back impurities were collected and repurified with the same strategy. Purity analysis:
Equipment: Kontron software KS2000, column: Waters Symmetry C18, 250 x 4.6 mm, 5μιη, 100 A. Flow rate: 1 ml/min, Temperature: 60°C, Wavelength: 210 nm Eluants: A : 0.1 % TFA in MilliQ water (1 ml trifluoracetic acid in 1 I of water); B : 0.1 % TFA in acetonitrile (1 ml trifluoracetic acid in 1 I of acetonitrile); Gradient : 20-80 % in 30 min. Injection volume: 20 μΙ of a solution at 0.5 mg/ml in 30% of acetonitrile.
Amino acid analysis:
Protocol: the peptide is first converted into its constituent amino acids by hydrolysis in constant boiling propionic acid, at 1 10°C, for 20 hours. Determination of amino acids, based on reversed- phase HPLC, is then performed according to the precolumn method derivatization with OPA reagent for primary amines and Fmoc-CI reagent for secondary amines with UV detection at 338 nm and 262 nm. Results were in agreement with the proposed amino acid composition.
Conjugation Methods
Method A: thiol-bromide coupling
Pentasaccharide 5 or 8 was dissolved in DMF (10 mg/mL) and hydroxylamine was added (25 equiv.). The solution was stirred for 10 minutes and was subsequently added to a solution of peptide P1 in DMF (3 mg/mL). EDTA, disodium salt (5 equiv.) was added and the mixture was stirred overnight at room temperature under a nitrogen atmosphere. The solution was diluted with water (3x) and purified by ion exchange chromatography on a Q Sepharose (Q FF HiTrap™ 5 mL) column). Fractions were analyzed with MonoQ ion exchange chromatography. Appropriate fractions containing conjugated kisspeptide were collected and purified by hydrophobic interaction chromatography (HIC Phenyl FF HiTrap™ high sub 5 mL column) using 3M NaCI as eluent. The combined fractions containing the conjugate were desalted by gel filtration chromatography on a G-25 sephadex column using water. Appropriate fractions were collected, filtered over a sterile 0.22 μιη filter and lyophilized to yield the conjugate as a white solid.
Method B: squarate coupling
Boric acid (3.09 g), KCI (2.61 g) and KOH (0.84 g) were dissolved in 100 mL water and the pH was adjusted to 9.0 by addition of solid KOH. Subsequently, peptide P2 or P3 was dissolved in a 1 : 1 mixture of MeOH and the aforementioned borate buffer (5 mg/mL). Next, pentasaccharide 3 or 4 (1.5 equiv.) was added as a solution water (30 mg/mL). After 1 hour solids started to appear in the reaction mixture and ACN was added until a clear solution was obtained. After overnight stirring the organic solvents were removed by evaporation under vacuum and water was added.
Purification of the conjugate was performed as described in Method A.
Method C: coupling by a cycloaddition reaction
Compound P4 (0.50 g, 0.33 mmol) was dissolved in DMF (20 mL). NMM (0.054 mL, 0.494 mmol, 1.5 eq) was added under a nitrogen atmosphere, followed by a solution of pentasaccharide 7 (0.65 g, 0.33 mmol) in water (10 mL). Nitrogen was bubbled through the solution for 5 min. Then sodium ascorbate (32.7 mg, 0.165 mmol, 0.5 equiv.) was added, followed by TBTA (17.5 mg, 0.165 mmol, 0.5 eq) and copper sulfate (26.3 mg, 0.165 mmol, 0.5 eq). The solution was stirred 30 minutes at r.t. A solution of EDTA disodium salt (380 mg, 1.13 mmol) in water (10 mL) was added and stirred for 30 minutes before drop wise addition of ACN (200 mL). After stirring for 20 minutes the precipitate was filtered off and washed with ACN. The residue was dried for 30 minutes before flushed with water (200 mL) and captured in a clean flask. The solution was eluted on a RP18 lichroprep column (70 mL) followed by water (600 mL). Then the column was eluted with a mixture of ACN and water (1/9, 1800 mL) and captured in fraction of 20 mL. The fractions containing product were collected and lyophilized to yield compound C8 (608 mg, 0.178 mmol, 53.8%).
This method is also representative for conjugation of kisspeptide derivatives P5-P27 to pentasaccharides 6, 7, 9 and 10. Table C: Kisspeptide-pentasaccharide conjugates:
Conjugation methodology and analytical data
Figure imgf000031_0001
P1 (2.2 mg, 1.5 umol, 1 equiv.) was conjugated to pentasaccharide 5 (4.3 mg, 2.0 umol, 1.3 equiv.) according to method A. Yield: 1.1 mg (21 %).
Conjugate C2
Figure imgf000032_0002
P1 (1 .9 mg, 1.3 umol, 1 equiv.) was conjugated to pentasaccharide 8 (3.4 mg, 1.7 umol, 1.3 equiv.) according to method A. Yield: 0.5 mg (10%). Theoretical mass: 3348. MS results: MIM= 3148 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M-S03]n +).
Conjugate C3
Figure imgf000033_0001
Peptide P2 (5.6 mg, 3.9 umol, 1 equiv.) was conjugated to pentasaccharide 4 (8.8 mg, 4.2 mmol, 1.06 equiv.) according to method B. Yield: 3.5 mg (26%). Theoretical mass: 3531. MS results: ammonium adducts of MIM 3330.9 M-9 Na).
Figure imgf000033_0002
Peptide P2 (5.5 mg, 3.8 umol, 1 equiv.) was conjugated to pentasaccharide 2 (7.2 mg, 3.8 mmol, 1 equiv) according to method B. Yield: 1 .5 mg (12%). Theoretical mass: 3298. MS results:
ammonium adducts of MIM 3087(M-9 Na).
Figure imgf000034_0001
Peptide P3 (2 mg, 1.18 umol, 1 equiv.) was conjugated to pentasaccharide 2 (3 mg, 1.6 umol, 1.35 e uiv.) according to method B. Yield: 1.7 mg (41 %).
Figure imgf000034_0002
Peptide P4 (3.2 mg, 2.2 mmol, 1 equiv.) was conjugated to pentasaccharide 10 (5.8 mg, 2.6 mmol, 1.2 equiv.) according to method C. Yield: 2 mg (26%). Theoretical mass: 3658. MS results: ammonium adducts of MIM 3458(M- 9 Na). Conjugate C7
Figure imgf000035_0001
Peptide P4 (32 mg, 0.019 mmol, 1 equiv.) was conjugated to pentasaccharide 6(77 mg, 0.041 mmol, 2 equiv.) according to method C. Yield: 15 mg (24%). Theoretical mass: 3337. MS results: ammonium adducts of MIM 3158.9 (M-8 Na)
Conjugate C8
Figure imgf000035_0002
Peptide P4 (50 mg, 0.033 mmol, 1 equiv.) was conjugated to pentasaccharide 7 (65 mg, 0.033 mmol, 1.0 equiv.) according to method C. Yield: 61 mg (54%). Theoretical mass: 3224.8.
Figure imgf000036_0001
Peptide P5 (3.5 mg, 2.2 mmol, 1 equiv.) was conjugated to pentasaccharide 7 (5.8 mg, 2.6 mmol, 1.2 equiv.) according to method C. Yield: 1 mg (12%). Theoretical mass: 3792.5. MS results: ammonium adducts of MIM 3592 M- 9 Na).
Figure imgf000036_0002
Peptide P6 (43.3 mg, 27 umol, 1 equiv.) was conjugated to pentasaccharide 6 (70.0 mg, 41 umol, 1.5 equiv) according to method C. Yield: 3.0 mg (10%). Theoretical mass: 3485. MS results: MIM 3307.
Figure imgf000037_0001
Peptide P6 (20.6 mg, 13 umol, 1 equiv.) was conjugated to pentasaccharide 7 (25.2 mg, 13 umol, 1.0 e uiv.) according to method C. Yield: 24.1 mg (56%).
Figure imgf000037_0002
Peptide P7 (5.9 mg, 3.8 umol, 1 equiv.) was conjugated to pentasaccharide 6 (9.8 mg, 5.7 umol, 1.5 equiv.) according to method C. Yield: 5.4 mg (41 %).
Figure imgf000038_0001
Peptide P7 (5.5 mg, 3.6 umol, 1 equiv.) was conjugated to pentasaccharide 7 (9.8 mg, 5.5 umol, 1.5 equiv.) according to method C. Yield: 3.5 mg (29%).
Conjugate C14
Figure imgf000038_0002
Peptide P8 (17.7 mg, 0.012 mmol, 1 equiv.) was conjugated to pentasaccharide 7 (32.2 mg, 0.018 mmol, 1.5 equiv.) according to method C. Yield: 1 1 .9 mg (29%). MS results: MIM= 3223
Figure imgf000039_0001
Peptide P9 (8.75 mg, 5.5 umol, 1 equiv.) was conjugated to pentasaccharide 7(11.3 mg, 5.7 umol, 1.05 equiv.) according to method C. Yield: 3.3 mg (18%).
Conjugate C16
Figure imgf000039_0002
umo, . 5 equv. accor ng to method C. Yield: 2.9 mg (16 ).
Figure imgf000040_0001
Peptide P10 (5.6 mg, 3.8 umol, 1 equiv.) was conjugated to pentasaccharide 7 (1 1.2 mg, 5.7 umol, 1.5 equiv.) according to method C. Yield: 4.5 mg (35%). Theoretical mass: 3449. MS results: MIM = 3248.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03 n+). Accurate mass and isotope fit are in agreement with structural formula.
Figure imgf000040_0002
Peptide P11 (18.7 mg, 0.013 mmol, 1 equiv) was conjugated to pentasaccharide 6 (47.5 mg, 0.025 mmol, 2 equiv.) according to method C. Yield: 26 mg (61 %). Theoretical mass: 3360. MS results: ammonium adducts of MIM 3181.98 (M-8Na).
Figure imgf000041_0001
Peptide P12 (10 mg, 6.1 umol, 1 equiv.) was conjugated to pentasaccharide 6 (1 1.6 mg, 6.8 umol, 1.1 equiv.) according to method C. Yield: 3.04 mg (15%).
Figure imgf000041_0002
umol, 1.1 equiv.) according to method C. Yield: 4.16 mg (20%). Conjugate C21
Figure imgf000042_0001
Peptide P13 (4.7 mg, 3.1 umol, 1 equiv.) was conjugated to pentasaccharide 7 (9.2 mg, 4.7 umol, 1.5 equiv.) according to method C. Yield: 2.6 mg (24%). Theoretical mass: 3449. MS results: MIM = 3264 observed.
Figure imgf000042_0002
Peptide P14 (9.5 mg, 6 umol, 1 equiv.) was conjugated to pentasaccharide 7 (14.3 mg, 7.3 umol, 1.2 equiv.) according to method C. Yield: 15.2 mg (73%). Theoretical mass: 3425. MS results: MIM = 3224.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M-S03]n+). Accurate mass and isotope fit are in agreement with structural formula. Conjugate C23
Figure imgf000043_0001
Peptide P15 (3.8 mg, 2.4 mmol, 1 equiv.) was conjugated to pentasaccharide 7 (5.6 mg, 2.9 mmol, 1.2 equiv.) according to method C. Yield: 2.4 mg (28%). Theoretical mass: 3559. MS results: ammonium adducts of MIM 3359 M-9 Na).
Figure imgf000043_0002
Peptide P16 (4.7 mg, 3.1 umol, 1 equiv.) was conjugated to pentasaccharide 7 (9.2 mg, 4.7 umol, 1.5 equiv.) according to method C. Yield: 7.2 mg (71 %).
Figure imgf000044_0001
Peptide P17 (1 1.6 mg, 7.29 umol, 1 equiv.) was conjugated to pentasaccharide 6 (20.6 mg, 10.9 umol, 1.5 equiv.) according to method C. Yield: 8.62 mg (35%). Theoretical mass: 341 1. MS results: MIM = 3232.95. observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03 n+). Accurate mass and isotope fit are in agreement with structural formula.
Figure imgf000044_0002
Peptide P17 (1 1.2 mg, 7.29 umol, 1 equiv.) was conjugated to pentasaccharide 7 (12.5 mg, 10.9 umol, 1.5 equiv.) according to method C. Yield: 8.58 mg (36%). Theoretical mass: 3499. MS results: MIM = 3298.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03]n+). Accurate mass and isotope fit are in agreement with structural formula.
Figure imgf000045_0001
Peptide P18 (8 mg, 5.24 umol, 1 equiv.) was conjugated to pentasaccharide 7 (15.5 mg, 7.9 umol, 1.5 equiv.) according to method C. Yield: 7.8 mg (50%). Theoretical mass: 3266.9. MS results: MIM = 3066.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03]n+). Accurate mass and isotope fit are in agreement with structural formula.
Conjugate C28
Figure imgf000046_0001
Peptide P19 (8.5 mg, 5.12 umol, 1 equiv.) was conjugated to pentasaccharide 7 (15.1 mg, 7.7 umol, 1.5 equiv.) according to method C. Yield: 5.8 mg (34%). Theoretical mass: 3514.2. MS results: MIM = 3313.9 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03]n+). Accurate mass and isotope fit are in agreement with structural formula.
Figure imgf000047_0001
Peptide P20 (5.8 mg, 4.08 umol, 1 equiv.) was conjugated to pentasaccharide 7 (12.1 mg, 6.12 umol, 1.5 equiv.) according to method C. Yield: 5.86 mg (46%). Theoretical mass: 3384.9. MS results: MIM = 3184.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03]n+). Accurate mass and isotope fit are in agreement with structural formula.
Figure imgf000047_0002
Peptide P22 (9.46 mg, 7.23 umol, 1 equiv.) was conjugated to pentasaccharide 6 (13.6 mg, 7.23 umol, 1.0 equiv.) according to method C. Yield: 5.42 mg (23%).
Figure imgf000048_0001
Peptide P22 (45.8 mg, 35 umol, 1 equiv.) was conjugated to pentasaccharide 7 (103 mg, 53 umol, 1.5 equiv.) according to method C. Yield: 93 mg (76%). MIM =3067.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M-S03]n+). Accurate mass and isotope fit are in agreement with structural formula.
Conjugate C32
Figure imgf000048_0002
Peptide P23 (23.3 mg, 16 umol, 1 equiv.) was conjugated to pentasaccharide 9 (51 .3 mg, 24 umol, 1.5 equiv.) according to method C. Yield: 14.7 mg (26%). Theoretical mass: 3570. MS results: MIM 3392 (product without sodium-salts) observed, no [M+H], but ammonium adducts observed
Figure imgf000049_0001
Peptide P23 (4.8 mg, 3.3 umol, 1 equiv.) was conjugated to pentasaccharide 10 (10.0 mg, 5.0 umol, 1.5 equiv.) according to method C. Yield: 5.5 mg (45%). Theoretical mass: 3658. MS results: MIM 3458 (product without sodium-salts) observed, no [M+H], but ammonium adducts observed
Figure imgf000049_0002
Peptide P23 (20.6 mg, 0.014 mmol, 1 equiv.) was conjugated to pentasaccharide 6 (40 mg, 0.021 mmol, 1.5 equiv.) according to method C. Yield: 30 mg (63%). Theoretical mass: 3337. MS results: ammonium adducts of MIM 3158.9 (M-8 Na).
Figure imgf000050_0001
Peptide P23 (20.6 mg, 0.014 mmol, 1 equiv.) was conjugated to pentasaccharide 7 (42 mg, 0.021 mmol, 1.5 equiv.) according to method C. Yield: 25 mg (52%). Theoretical mass: 3425. MS results: ammonium adducts of MIM 3224.8 (M-9Na).
Conjugate C36
Figure imgf000050_0002
Peptide P24 (4.3 mg, 3.3 umol, 1 equiv.) was conjugated to pentasaccharide 6 (9.3 mg, 4.94 umol, 1.5 equiv.) according to method C. Yield: 4.0 mg (38%). Conjugate C37
Figure imgf000051_0001
Peptide P24 (4.3 mg, 3.3 umol, 1 equiv.) was conjugated to pentasaccharide 7 (9.7 mg, 4.94 umol, 1.5 equiv.) according to method C. Yield: 2.2 mg (21 %).
Conjugate C38
Figure imgf000051_0002
Peptide P25 (10.1 mg, 7.8 umol, 1 equiv.) was conjugated to pentasaccharide 6 (15.3 mg, 8.2 umol, 1.05 equiv.) according to method C. Yield: 12.8 mg (55%). Conjugate C39
Figure imgf000052_0001
Peptide P25 (10.1 mg, 7.8 umol, 1 equiv.) was conjugated to pentasaccharide 7 (16.1 mg, 8.2 umol, 1.05 e uiv.) according to method C. Yield: 15.2 mg (64%).
Figure imgf000052_0002
Peptide P26 (2.58 mg, 1.79 umol, 1 equiv.) was conjugated to pentasaccharide 6 (3.53 mg, 1.88 umol, 1.05 equiv.) according to method C. Yield: 1.61 mg (29%).
Figure imgf000053_0001
Peptide P27 (1.35 mg, 0.91 umol, 1 equiv.) was conjugated to pentasaccharide 6 (3.1 mg, 1.8 umol, 1.9 equiv.) according to method C. Yield: 0.96 mg (31 %).
Figure imgf000053_0002
Peptide P21 (1 1.7 mg, 8.19 umol, 1 equiv.) was conjugated to pentasaccharide 7 (24.2 mg, 12 umol, 1.5 equiv.) according to method C. Yield: 16.3 mg (58%). Theoretical mass: 3396. MS results: MIM = 3195.8 observed in the form of ammonium adducts for (M2+,M3+,M4+ and [M- S03]n+). Accurate mass and isotope fit are in agreement with structural formula. Biological activity
In vitro agonist activity at the KISS-receptor.
The in vitro agonistic activity of the kisspeptide-pentasaccharide conjugates of the invention at the Gaq-coupled human GPR54-R was measured in the absence and presence of 20% human serum in a NFAT-regulated luciferase reporter gene assays. CHO-k1 cells were used that expressed the mitochondrially targeted aequorine and Ga16 (from Euroscreen, Brussels, Belgium), a Nuclear Factor of Activated T-cells-responsive element (NFAT) under the control of a promoter directing the expression of a firefly luciferase reporter gene (pNFAT-TALuc obtained from Clontech, Mountain View, CA), and the human GPR54-R (pcDNA3.1 Hygro(+)- hGPR54WT c4; MSD). These cells were cultured in Dulbecco's Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F12) (Invitrogen, Carlsbad, CA), supplemented with 10 % (vol/vol) filtered bovine serum (Hyclone, Thermo Fisher Scientific, Waltham, MA), 100 U/ml penicillin G, 100 μg/ml streptomycin, 400 μg/ml Geneticin and 400 μg/ml Hygromycin (Invitrogen). Kisspeptide- pentasaccharide conjugates and Kisspeptin-10 (Metastin 45-54 from Pepnet; Louisville, KY) were diluted in assay medium (DMEM/F12 containing 1 μg/ml insulin (Diosynth, Oss, The
Netherlands), 5 μg/ml apo-transferin (Sigma-Aldrich, St. Louis, MO) and 0.1 % (vol/vol) bovine serum albumine (BSA, Sigma Aldrich)) in a 10-point serial dilution. Kisspeptin-10 was used as a standard. 10 μ I of kisspeptide-pentasaccharide conjugate and Kisspeptin-10 dilutions were added to a white 384-wells culture plate (Perkin Elmer, Boston, MA). When the conjugates were tested in the presence of a final percentage of 20% human serum, 10 μΙ assay medium containing 60% human serum (Sigma-Aldrich, #H4522) was added to the wells. In the absence of human serum, 10 μΙ of assay medium was added. Cells were harvested and diluted to a concentration of 7.5E5 cells/ml in assay medium without 0.1 % BSA. 10 μΙ of cell solution was added to the compound wells. After incubation for 4 hours in a humidified atmosphere at 5% C02 and 37 °C, the plate was left at room temperature for 30 minutes. Subsequently, 15 μ I of Steadylite™ solution
(PerkinElmer, Boston, MA) was added and the cells, protected from light, were allowed to lyse for 30 min at room temperature. Luminescence, indicated as relative luminescence units (RLU), was measured on an Envision Multilabel reader (PerkinElmer).
The effect of kisspeptide-pentasaccharide conjugate was calculated as percentage of the maximum effect of Kisspeptin-10 (effect at 1 E-6M) and the minimum effect (assay medium).
Curves were fit by nonlinear regression using the sigmoidal dose-response equation in GraphPad Prism version 4.03 (GraphPad, San Diego, CA), resulting in a pEC5o value for the conjugate
(Table I). In vitro stability study of kisspeptide-pentasaccharide conjugates in human serum.
Test compound is added to 1.5 mL human serum in a 96-wells deepwell plate in a final concentration of 750 nM. The compound is mixed and immediately thereafter a 100 μΙ sample is harvested in a 96-wells 2D-tuberack (T=0 sample). The 2 D-tubes are closed with rubber tube caps and the sample plate is stored at -20°CF in the freezer. The deepwell plate is put into an incubator which was set at humidified (95%) atmosphere at 5-7% C02 and 37°C for an incubation period of maximum 240h. During this incubation period, samples of 100 μΙ are collected on various time intervals, dependent on the test compound used; time points 0, 1 , 2, 3, 6, 24, 48, 72h in case of Kisspeptides and 0, 3, 6, 24, 48, 72, 168, 240h in case of kisspeptide- pentasaccharide conjugates. Sample plates are put on ice in case of sampling and stored at -20°C until analysis by LC/MS/MS. Determination of kisspeptide-pentasaccharide conjugates in animal and human plasma by LC- MS/MS
Kisspeptide-pentasaccharide conjugates are determined by LC-MS/MS after extraction from 20 or 100 μΙ_ plasma samples by protein precipitation. Protein precipitation is executed after the addition of heparin and by common acetonitrile precipitation. For LC-MS/MS analysis, the compounds are separated over an XTerra RP8 column and using a gradient mobile phase of ammonium acetate buffer and acetonitrile at a fixed column temperature. MS/MS detection is done with a triple quadrupole mass spectrometer and employing negative ion electrospray ionization. The mass spectrometer is set up to achieve a known degree of in source
fragmentation, where the initially formed multiply charged negative ions lose one or more S03 molecules. The most abundant of these secondary ions are then subjected to a common MS/MS experiment in which a single SO3 loss is monitored. The Q1 and Q3 mass filter settings for this MS/MS experiment have commonly been determined in method development. The Q1 and Q3 masses are highly selective for each individual kisspeptide-pentasaccharide conjugate. Internal standardization is accomplished by adding a different conjugate of approximately the same size to the bioanalytical samples. Although the limit of quantitation may vary for the individual kisspeptide-pentasaccharide conjugates, the observed sensitivity generally supports an assay range of 0.1 nM to 2000 nM sample concentrations.
The obtained concentration data are analyzed from the plasma concentration vs time curve by means of the Non Compartmental Analysis (NCA) module in WinNonlin (Pharsight). The rate of compound loss is expressed as a half-life (plasma t1/2). The t1/2 for the test compound can be determined by plotting the natural logarithm (In) of the measured concentrations against time and fitting the line through all data points. The gradient of this line is the first order rate constant (k) for substrate disappearance and is determined by regression analysis. The rate constant is converted to a half-life according to the following equation:
In vitro half-life (t1/2) = ng Table I. Agonist potencies of kisspeptide-pentasaccharide conjugates of the invention at the human KISS-receptor in the absence and presence of 20% serum in a NFAT-luciferase
Figure imgf000056_0001
In vivo pharmacokinetic study in rat.
The pharmacokinetics of conjugate C8 were determined after administration of 40 nmol/kg IV in male and of 200 nmol/kg SC in female Sprague-Dawley rats. The measured PK parameters (Tables II and I II) show that the plasma clearance of the conjugate was very low (0.08-0.012 mL/min/kg) and that the distribution volume in steady state is limited to the blood volume.
The terminal elimination half-life for conjugate C8 was 1 1 .5 hr in rat. Following subcutaneous administration, absorption was rapid (tmax 2.7 - 3.3 hr) and % bioavailability was 94 in rat (Table II).
Table II . Pharmacokinetics in rats
Figure imgf000057_0001
AUCN = dose-normalized area under the plasma concentration-time curve; Clp= plasma clearance; Vd^ volume of distribution at steady state; F= bioavailability. Numbers represent average values from 3 animals per tested treatment and species.
Conjugate C8 was dissolved in 0.1 % BSA in saline for IV administration in rats, and in 5% mannitol in 6 mM acetate buffer pH 5.0 for SC administration in female rats.
Table III. SC Pharmacokinetics in rats
Figure imgf000057_0002
The results disclosed in Table I , I I and I II reveal that the conjugates of the present invention were surprisingly found to be highly potent (pEC50 >8 in the presence of human serum) and sufficiently stable (T1/2 >24h in human serum) to be applied as long acting kisspeptide agonist for treatment of female infertility. Moreover, it was corroborated that the conjugates of the present invention were fully stable in the blood circulation in vivo (i.e. the kisspeptide- pentasaccharide conjugate was not prone to proteolytic degradation and it was excreted as the intact molecule). In contrast, the reference peptides P28 (kisspeptin-10) and P29 (see Table A and Table I) were not sufficiently stable (and they are still prone to fast excretion in vivo due to their small molecular size), while reference conjugate C42 (kisspeptin-10 pentasaccharide conjugate) is not sufficiently potent.

Claims

Claims.
A kisspeptide-pentasaccharide conjugate having the formula (I)
Ri -Zi -(Asn)n-Z3-Asn-Z5-Phe-Z7-Z8-Arg(R3)-Zi 0-NH2
Formula (I)
wherein
! is Tyr or D-Tyr;
Z3 is Trp, Hyp, Phe or Lys(R2) ;
Z5 is Thr, Aib or Ala;
Z7 is Gly or azaGly;
Z8 is Leu; or
Z7 and Z8 together represent
Figure imgf000059_0001
Z10 is Phe or Trp;
n is 0 or 1 ;
Ri or R2, when present, represents a pentasaccharide derivative having the formula (I I)
Figure imgf000059_0002
Formula (I I);
wherein R is methyl or S03X;
X is a positively charged counterion;
with the proviso that when R2 is present, is H or (d-e) alkylcarbonyl;
R3 is H or (Ci-3)alkyl; and
L represents a pharmacologically inactive linker moiety having 1 0-50 atoms; or a pharmaceutically acceptable salt thereof.
2. The kisspeptide-pentasaccharide conjugate of claim 1 , wherein
Z5 is Thr.
3. The kisspeptide-pentasaccharide conjugate of claim 1 or 2, wherein
R3 is methyl.
4. The kisspeptide-pentasaccharide conjugate of any one of claims 1 -3, wherein n is 0; 7- is D-Tyr; Z3 is Hyp and Z10 is Trp.
5. The kisspeptide-pentasaccharide conjugate of any one of claims 1 -3, wherein n is 1 and Z10 is Phe.
6. The kisspeptide-pentasaccharide conjugate of any one of claims 1 -5, wherein R is methyl.
7. The kissp
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000061_0001
59
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000063_0002
61
Figure imgf000064_0001
or a pharmaceutically acceptable salt thereof.
The kisspeptide-pentasaccharide conjugate of any one of claims 1 -7 for use in therapy. The use of a kisspeptide-pentasaccharide conjugate of any one of claims 1 -7 in the treatment of female infertility.
A pharmaceutical composition comprising a kisspeptide-pentasaccharide conjugate of any one of claims 1 -7 or a pharmaceutically acceptable salt thereof, in admixture with pharmaceutically acceptable auxiliaries
PCT/EP2012/065061 2011-08-04 2012-08-01 Kisspeptide-pentasaccharide conjugates WO2013017631A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/236,694 US9127038B2 (en) 2011-08-04 2012-08-01 Kisspeptide-pentasaccharide conjugates
EP12743963.6A EP2755690B1 (en) 2011-08-04 2012-08-01 Kisspeptide-pentasaccharide conjugates

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11176518 2011-08-04
EP11176518.6 2011-08-04

Publications (1)

Publication Number Publication Date
WO2013017631A1 true WO2013017631A1 (en) 2013-02-07

Family

ID=46639489

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/065061 WO2013017631A1 (en) 2011-08-04 2012-08-01 Kisspeptide-pentasaccharide conjugates

Country Status (3)

Country Link
US (1) US9127038B2 (en)
EP (1) EP2755690B1 (en)
WO (1) WO2013017631A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR3001455A1 (en) * 2013-01-31 2014-08-01 Agronomique Inst Nat Rech AGONIST COMPOUNDS OF THE KISS1R RECEPTOR AND THEIR USE IN INDUCING OVULATION IN MAMMALS
JP2017524768A (en) * 2014-08-01 2017-08-31 マサチューセッツ インスティテュート オブ テクノロジー Modified alginate for antifibrotic materials and uses
JP2020029479A (en) * 2018-08-20 2020-02-27 新中村化学工業株式会社 Polymer compound and polymer composition containing the same, and inorganic particle-containing composition
JP2021518361A (en) * 2018-03-16 2021-08-02 サレプタ セラピューティクス, インコーポレイテッド Chimeric peptide for antisense delivery
US20230045718A1 (en) * 2016-09-30 2023-02-09 Myovant Sciences Gmbh Methods of increasing endogenous luteinizing hormone

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2571896C2 (en) * 2015-01-28 2015-12-27 "Частное учреждение образовательная организация высшего образования "Медицинский университет "РЕАВИЗ" Method for simulating female anovulatory infertility in rats

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1277761A2 (en) 2001-07-19 2003-01-22 Akzo Nobel N.V. Process for the preparation of peptides
WO2004063221A1 (en) 2002-12-26 2004-07-29 Takeda Pharmaceutical Company Limited Metastin derivative and use thereof
WO2006001499A2 (en) 2004-06-25 2006-01-05 Takeda Pharmaceutical Company Limited Metastin derivatives and use thereof
WO2006082184A2 (en) 2005-02-01 2006-08-10 N.V. Organon Conjugates of a polypeptide and a pentasaccharide
WO2007072997A1 (en) 2005-12-22 2007-06-28 Takeda Pharmaceutical Company Limited Metastin derivatives and use thereof
WO2008029278A2 (en) 2006-08-03 2008-03-13 Endotis Pharma S.A. Conjugates of antithrombin binding oligosaccharide derivatives and therapeutic proteins
WO2010013762A1 (en) 2008-07-30 2010-02-04 武田薬品工業株式会社 Metastin derivative and use thereof
WO2010033224A1 (en) 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of kiss1 peptides
WO2010033207A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of therapeutic peptides

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2562676A1 (en) 2004-04-23 2005-12-15 Applied Research Systems Ars Holding N.V. Use of gpcr54 ligands for the treatment of infertility
TWI404726B (en) 2006-10-25 2013-08-11 Takeda Pharmaceutical Metastin derivatives and use thereof

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1277761A2 (en) 2001-07-19 2003-01-22 Akzo Nobel N.V. Process for the preparation of peptides
WO2004063221A1 (en) 2002-12-26 2004-07-29 Takeda Pharmaceutical Company Limited Metastin derivative and use thereof
WO2006001499A2 (en) 2004-06-25 2006-01-05 Takeda Pharmaceutical Company Limited Metastin derivatives and use thereof
WO2006082184A2 (en) 2005-02-01 2006-08-10 N.V. Organon Conjugates of a polypeptide and a pentasaccharide
WO2007072997A1 (en) 2005-12-22 2007-06-28 Takeda Pharmaceutical Company Limited Metastin derivatives and use thereof
WO2008029278A2 (en) 2006-08-03 2008-03-13 Endotis Pharma S.A. Conjugates of antithrombin binding oligosaccharide derivatives and therapeutic proteins
WO2010013762A1 (en) 2008-07-30 2010-02-04 武田薬品工業株式会社 Metastin derivative and use thereof
EP2314609A1 (en) * 2008-07-30 2011-04-27 Takeda Pharmaceutical Company Limited Metastin derivative and use thereof
WO2010033224A1 (en) 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of kiss1 peptides
WO2010033207A1 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Polymer conjugates of therapeutic peptides

Non-Patent Citations (58)

* Cited by examiner, † Cited by third party
Title
"Peptide Synthesis, Structures, and Applications", 1995, ACADEMIC PRESS
"Peptides: Chemistry and Biology", 2002, WILEY-VCH
"The Peptides, Analysis, Synthesis, Biology", vol. 3, 1981, ACADEMIC PRESS
A. BRIK ET AL., CHEM. BIOCHEM, vol. 6, 2005, pages 1167 - 1169
BODANSZKY: "Principles of Peptide Synthesis", 1984, SPRINGER-VERLAG
BUIJSMAN R C ET AL: "Design and synthesis of a novel synthetic NAPAP-penta-saccharide conjugate displaying a dual antithrombotic action", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 9, no. 14, 19 July 1999 (1999-07-19), pages 2013 - 2018, XP004171628, ISSN: 0960-894X, DOI: 10.1016/S0960-894X(99)00320-0 *
BUIJSMAN R C ET AL: "Synthesis of a pentasaccharide-oligodeoxyribonucleotide conjugate : a novel antithrombotic agent", CHEMISTRY - A EUROPEAN JOURNAL, vol. 2, no. 12, 1 January 1996 (1996-01-01), pages 1572 - 1577, XP002363505, ISSN: 0947-6539, DOI: 10.1002/CHEM.19960021216 *
C. CHEN ET AL., BIORG. MED. CHEM. LETT., vol. 19, 2009, pages 3875 - 3879
CASSUTT KJ ET AL., J OF BIOMOL SCREENING, vol. 12, 2007, pages 285 - 287
CHANNA N. JAYASENA ET AL: "Kisspeptin: Paving the Way to a New Therapeutic Avenue in Reproduction", RECENT PATENTS ON ENDOCRINE, METABOLIC & IMMUNE DRUG DISCOVERY, vol. 3, no. 2, 1 June 2009 (2009-06-01), pages 87 - 93, XP055022221, ISSN: 1872-2148, DOI: 10.2174/187221409788452354 *
CHRISTOF M. NIEMEYER: "Bioconjugation Protocols", vol. 283, 2004, HUMANA PRESS
D. CRICH: "Reagents for Glycoside, Nucleotide and Peptide Synthesis", 2005, WILEY
DE KORT ET AL.: "Conjugation of ATIII-binding pentasaccharides to extend the half-life of proteins: long- acting insulin", CHEMMEDCHEM, vol. 3, 2008, pages 1189 - 1193
DHILLO W ET AL., J CLIN ENDOCRINOL METAB, vol. 92, 2007, pages 3958 - 3966
GENNARO, A.R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WILKINS
GOTTSCH ET AL., PEPTIDES, vol. 30, 2009, pages 4 - 9
GREG T. HERMANSON: "Bioconjugate Techniques", 2008, ACADEMIC PRESS
H. AHMED: "Principles and Reactions of Protein Extraction, Purification and Characterization", 2005, CRC PRESS
HANSEN KB; BRAUNER-OSBORNE H, METHODS MOL BIO, vol. 1552, 2009, pages 269 - 78
HUET ET AL.: "Long-lasting enfuvirtide carrier pentasaccharide conjugates with potent anti-human immunodeficiency virus type 1 activity", ANTIMICROB. AGENTS CHEMOTHER, vol. 54, 2010, pages 134 - 142, XP055125299, DOI: doi:10.1128/AAC.00827-09
I.F. EGGEN ET AL., J. PEPTIDE SCI., vol. 11, 2005, pages 633 - 641
I.F. EGGEN ET AL., ORG. PROC. RES. DEV., vol. 9, 2005, pages 98 - 101
J. ORG. CHEM., vol. 64, 1999, pages 7388
J. ORG. CHEM., vol. 72, 2007, pages 7963 - 7967
J. SPRINGER ET AL., EUR. J. ORG. CHEM., vol. 15, 2008, pages 2592 - 2600
JEAN-FRANCOIS LUTZ: "1,3-Dipolar Cycloadditions of Azides and Alkynes A Universal Ligation Tool in Polymer and Materials", ANGEW. CHEM. INT. ED., vol. 46, 2007, pages 1018 - 1025, XP002500213, DOI: doi:10.1002/ANIE.200604050
JIA ET AL., MOL ENDOCRINO, vol. 5, 1991, pages 759 - 776
K.L. KIICK ET AL., PROC. NAT. ACAD. SCI., vol. 99, 2002, pages 19 - 24
K.M. GOODING; F.E. REGNIER: "HPLC of Biological Macromolecules", 2002, MARCEL DEKKER INC.
KNOP ET AL.: "Poly(ethylene glycol) in drug delivery: pros and cons as well as potential alternatives", ANGEW. CHEM. INT. ED., vol. 49, 2010, pages 6288 - 6308, XP055251511, DOI: doi:10.1002/anie.200902672
LUTZ ET AL., BIOCONJUGATE CHEM., vol. 2, 1991, pages 148 - 153
M. LEWINSKA ET AL., BIOCONJUGATE CHEM., vol. 15, 2004, pages 231 - 234
M. SATO ET AL., J. AM. CHEM. SOC., vol. 126, 2004, pages 14013 - 14022
M.A. GAUTHIER ET AL.: "Peptidelprotein-polymer conjugates: synthetic strategies and design concepts", CHEM. COMMUN., 2008, pages 2591 - 2611, XP007910450, DOI: doi:10.1039/b719689j
M.F. DEBETS ET AL., CHEMBIOCHEM, vol. 11, 2010, pages 1168 - 1184
M.-I. AGUILAR: "HPLC of Peptides and Proteins, Methods and Protocols", 2004, HUMANA PRESS
MCCORMACK AL ET AL., ANAL. CHEM., vol. 69, no. 4, 1997, pages 767 - 776
MERRIFIELD, R. B., J. AM. CHEM. SOC., vol. 85, 1963, pages 2149 - 2154
MICHAEL D. BEST: "Click Chemistry and Bioorthogonal Reactions: Unprecedented Selectivity in the Labeling of Biological Molecules", BIOCHEMISTRY, vol. 48, 2009, pages 6571 - 6584, XP002659165, DOI: doi:10.1021/bi9007726
N. OLIVIER ET AL., BIOCONJUGATE CHEM., vol. 20, 2009, pages 1397 - 1403
N.L.BENOITON: "Chemistry of Peptide Synthesis", 2005, CRC TAYLOR & FRANCIS
ORG. LETT., vol. 3, 2001, pages 781 - 783
ORG. LETT., vol. 8, 2006, pages 4505 - 4507
ORG. LETT., vol. 9, 2007, pages 3797 - 3800
P. SCHELTE ET AL., BIOCONJUGATE CHEM., vol. 11, 2000, pages 118 - 123
PEP?IDES, vol. 5, no. 2, 1984, pages 167 - 168
ROA ET AL., ENDOCRINOLOGY, vol. 147, 2006, pages 2864 - 2878
ROGER L. LUNDBLAD: "Chemical Reagents for Protein Modification", 2005, CRC PRESS
S.I. PRESOLSKI ET AL., J. AM. CHEM. SOC., vol. 132, 2010, pages 14570 - 14576
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", COLD SPRING HARBOR LABORATORY PRESS
SEWALD, N.: "Peptides: Chemistry and Biology", 2003, WILEY-VCH
STRATOWA ET AL., CURR OPIN BIOTECHNOL, vol. 6, 1995, pages 574
TOMITA ET AL., J. MED. CHEM., vol. 51, no. 23, 2008, pages 7645 - 7649
VAN BOECKEL; PETITOU, ANGEW. CHEM. INTL. ED. ENGL., vol. 32, 1993, pages 1671 - 1690
VAN KOPPEN: "Generic Assays in G-protein- coupled Receptor Drug Discovery", DRUG DISCOVERY, vol. 7, 2010, pages 69 - 72
VOGEL G M T ET AL: "ANTITHROMBOTIC PROPERTIES OF A DIRECT THROMBIN INHIBITOR WITH A PROLONGED HALF LIFE AND AT-MEDIATED FACTOR XA INHIBITORY ACTIVITY", JOURNAL OF THROMBOSIS AND HAEMOSTASIS, vol. 1, no. 9, 1 September 2003 (2003-09-01), pages 1945 - 1954, XP008045748, ISSN: 1538-7933, DOI: 10.1046/J.1538-7836.2003.00351.X *
WHITE: "Fmoc Solid Phase Peptide Synthesis, A practical Approach", 2003, OXFORD UNIVERSITY PRESS
YANG B ET AL: "Goldfish kisspeptin: Molecular cloning, tissue distribution of transcript expression, and stimulatory effects on prolactin, growth hormone and luteinizing hormone secretion and gene expression via direct actions at the pituitary level", GENERAL AND COMPARATIVE ENDOCRINOLOGY, vol. 165, no. 1, 1 January 2010 (2010-01-01), pages 60 - 71, XP026761775, ISSN: 0016-6480, [retrieved on 20090606], DOI: 10.1016/J.YGCEN.2009.06.001 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR3001455A1 (en) * 2013-01-31 2014-08-01 Agronomique Inst Nat Rech AGONIST COMPOUNDS OF THE KISS1R RECEPTOR AND THEIR USE IN INDUCING OVULATION IN MAMMALS
WO2014118318A1 (en) * 2013-01-31 2014-08-07 Institut National De La Recherche Agronomique Kiss1r receptor agonist compounds and use thereof for inducing ovulation in mammals
US9884891B2 (en) 2013-01-31 2018-02-06 Institut National De La Recherche Agronomique KISS1R receptor agonist compounds and use thereof for inducing ovulation in mammals
JP2017524768A (en) * 2014-08-01 2017-08-31 マサチューセッツ インスティテュート オブ テクノロジー Modified alginate for antifibrotic materials and uses
JP2020023582A (en) * 2014-08-01 2020-02-13 マサチューセッツ インスティテュート オブ テクノロジー Modified alginates for anti-fibrotic materials and applications
US20230045718A1 (en) * 2016-09-30 2023-02-09 Myovant Sciences Gmbh Methods of increasing endogenous luteinizing hormone
JP2021518361A (en) * 2018-03-16 2021-08-02 サレプタ セラピューティクス, インコーポレイテッド Chimeric peptide for antisense delivery
JP2020029479A (en) * 2018-08-20 2020-02-27 新中村化学工業株式会社 Polymer compound and polymer composition containing the same, and inorganic particle-containing composition
JP7061795B2 (en) 2018-08-20 2022-05-02 新中村化学工業株式会社 Polymer compounds, polymer compositions containing them, and compositions containing inorganic particles.

Also Published As

Publication number Publication date
EP2755690A1 (en) 2014-07-23
US9127038B2 (en) 2015-09-08
EP2755690B1 (en) 2020-04-01
US20140206627A1 (en) 2014-07-24

Similar Documents

Publication Publication Date Title
AU2018211208B2 (en) Insulin receptor partial agonists
US11649269B2 (en) Bifunctional compounds comprising insulin peptides and EGF(A) peptides
EP2755690B1 (en) Kisspeptide-pentasaccharide conjugates
CN112074531A (en) GIP derivatives and uses thereof
BRPI0607248A2 (en) conjugate of a polypeptide and an oligosaccharide, pharmaceutical composition, use of the conjugate, and process for the preparation of a conjugate
WO2017023933A2 (en) Peptidomimetic macrocycles
JP2023540679A (en) Conjugated hepcidin mimetics
WO2022212698A1 (en) Conjugated hepcidin mimetics
KR20180038049A (en) Novel Insulin Derivatives and Their Medical Use
US20180022780A1 (en) Stabilization adrenomedullin derivatives and use thereof
US7273921B2 (en) Method for producing acylated peptides
US11975040B2 (en) Plexin binding regulator
EP3472195B1 (en) Metabolically stable spexin peptide analogs
US11612661B2 (en) Conotoxin peptide analogs and uses for the treatment of pain and inflammatory conditions
KR20230104118A (en) GLP-1 Prodrugs and Uses Thereof
CA3125383A1 (en) Conotoxin peptide analogs and uses for the treatment of pain and inflammatory conditions
CA3143584A1 (en) Adrenomedullin-analogues for long-term stabilization and their use
EP3936191A1 (en) Hemagglutinin-binding peptide
WO2024022465A1 (en) Human amylin polypeptide derivative and use thereof
WO2023137355A2 (en) Potent and stable polypeptide analogues via serine/threonine ligation
CN116457000A (en) Conjugated hepcidin mimetics
TW202026305A (en) Conotoxin peptide analogs and uses for the treatment of pain and inflammatory conditions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12743963

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012743963

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 14236694

Country of ref document: US