WO2008049866A1 - New chelating compound - Google Patents

New chelating compound Download PDF

Info

Publication number
WO2008049866A1
WO2008049866A1 PCT/EP2007/061438 EP2007061438W WO2008049866A1 WO 2008049866 A1 WO2008049866 A1 WO 2008049866A1 EP 2007061438 W EP2007061438 W EP 2007061438W WO 2008049866 A1 WO2008049866 A1 WO 2008049866A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound according
ser
xaa3
xaa1
xaa2
Prior art date
Application number
PCT/EP2007/061438
Other languages
French (fr)
Inventor
Anna Orlova
Vladimir Tolmachev
Original Assignee
Affibody Ab
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 Affibody Ab filed Critical Affibody Ab
Publication of WO2008049866A1 publication Critical patent/WO2008049866A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/081Tripeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0815Tripeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0819Tripeptides with the first amino acid being acidic

Definitions

  • the present invention relates to compounds useful for in vivo imaging. More particularly, the invention relates to a chelating compound and a chelate thereof with a radioactive metal atom. The present invention furthermore relates to methods of diagnosis, wherein the chelating compound is used.
  • Molecular targeting of chemical structures within the body of a subject has been evaluated, for example for purposes of in vivo diagnostics of cancer disease.
  • the targeting of tumor specific chemical structures is called tumor targeting.
  • Molecules used for detection in the various applications of molecular targeting are called targeting agents.
  • Monoclonal antibodies, fragments of monoclonal antibodies and peptides have been evaluated as targeting agents.
  • the large size of antibodies causes slow blood clearance, slow extravasation and slow tumor penetration. Therefore, the use of peptide receptor ligands for targeting of receptors that are overexpressed in malignant tumors is today a common approach in radionuclide tumor targeting. The small size of such peptides improves extravasation and tumor penetration.
  • US 4 861 869 discloses bifunctional coupling agents suitable for forming conjugates with biomolecules and for complexing with radionuclide atoms.
  • Examples of such coupling agents are various salts of modified mercaptoacetyl-glycyl-glycyl-glycinate, which are coupled to different antibody variants.
  • EP 284 071 A2 discloses similar labeling of biomolecules with a mercaptoacetyl-glycyl-glycyl-glycyl moiety for complexing with a radionuclide.
  • US 5 980 861 is directed to the labeling of polynucleotides via a chelator compound, which chelator compound is either mercaptoacetyl-glycyl- glycyl-glycyl or mercaptoacetyl-seryl-seryl-seryl.
  • EP 284 071 A2 is a schematic illustration of the geometry of such a complex.
  • N3S or N 3 S chelators are collectively denoted N3S or N 3 S chelators, due to the three N's and one S involved.
  • the work to date relating to such N3S chelators has been centered on mercaptoacetyl-glycyl-glycyl-glycyl-glycyl, which in the present text is denoted ma-Gly-Gly-Gly. It is also referred to in the literature as MAG3 or MAG3.
  • ma-Gly-Gly-Gly with different protective groups has been utilized for labeling of antibodies (Lei et al, Nucl Med Biol 23(7):917-922 (1996)) and their
  • N3S chelators were studied by Zhu et al in Nucl Med Biol 28:703-708 (2001 ). In order to develop and test an alternative synthesis route, the authors synthesized ma-Gly-Gly-Gly as well as glutamic acid or phenylalanine containing variants ma-Glu-Gly-Gly and ma-Phe-Gly-Gly. The effect on biodistribution in mice was studied via coupling of these chelators to the 7 kD HNE2 protein.
  • One object of the present invention is to meet this demand, by developing new and improved chelating compounds that can function as targeting agents when complexed to a suitable radioactive metal atom.
  • Another object of the present invention is to provide chelates of such compounds with radioactive metal atoms, which chelates show high sensitivity for target structures.
  • a related object is to provide chelates of such compounds with radioactive metal atoms, which chelates reduce the radioactivity accumulation in healthy tissue.
  • Yet another object of the present invention is to provide methods for preparing said chelates of compounds with radioactive metal atoms.
  • Still another object of the present invention is to provide means and methods that enable targeted in vivo imaging for diagnostic and/or pharmacokinetic studies.
  • - Xaa1 , Xaa2 and Xaa3 are amino acid residues defined as follows: o any one or two of Xaa1 , Xaa2 and Xaa3 is/are the amino acid residue Ser; o in case two of Xaa1 , Xaa2 and Xaa3 are Ser, the remaining amino acid residue is selected from the group of amino acid residues consisting of GIu, Arg and Lys; o in case one of Xaa1 , Xaa2 and Xaa3 is Ser, the remaining amino acid residues are both GIu residues.
  • - R has a biological function and is selected from the group consisting of polypeptides, PNA, DNA, RNA, and mixtures thereof.
  • the chelating compound according to the invention has a chelating moiety, ma-Xaa1 -Xaa2-Xaa3, and a moiety conferring a biological function, R.
  • a chelating moiety ma-Xaa1 -Xaa2-Xaa3
  • a moiety conferring a biological function, R a biological function
  • variants of the chelating moiety of the inventive chelating compounds are denoted ma-Xaa1 -Xaa2-Xaa3, each of said Xaa:s being an amino acid represented in the standard three-letter code.
  • a chelating moiety in a chelating compound according to the invention in which moiety Xaa1 is GIu, Xaa2 is Ser and Xaa3 is GIu, is thus denoted ma- Glu-Ser-Glu, GIu and Ser being the three letter representations of glutamic acid and serine, respectively.
  • the ma-Xaa1 -Xaa2-Xaa3 chelating moiety comprises one or two Ser, the remaining two or one amino acid residues being GIu.
  • Xaa3 may be GIu, and the inventive compound be selected from ma-Ser-Glu-Glu-R, ma-Glu-Ser-Glu-R and ma- Ser-Ser-Glu-R.
  • Xaa3 may be Ser, and the inventive compound be selected from ma-Ser-Glu-Ser-R, ma-Glu-Glu-Ser-R and ma-Glu-Ser-Ser- R.
  • ma-Xaa1 -Xaa2-Xaa3 comprises two Ser, the remaining amino acid residue being selected from Arg and Lys.
  • the remaining amino acid residue may be Arg, and the inventive compound be selected from ma-Ser-Ser-Arg-R, ma-Ser-Arg-Ser-R and ma-Arg-Ser-Ser- R.
  • the remaining amino acid residue may be Lys, and the inventive compound be selected from ma-Ser-Ser-Lys-R, ma-Ser-Lys-Ser-R and ma-Lys-Ser-Ser-R.
  • the inventive compound is selected from ma-Ser-Arg-Ser-R and ma-Ser-Lys-Ser-R.
  • the incorporation of one positively charged amino acid residue in the chelating moiety of the inventive compound unexpectedly provides the desired shift in excretion from the hepatobiliary pathway to the renal pathway.
  • renal uptake increased only marginally.
  • a chelating moiety having three positively charged amino acids e.g. ma-Lys-Lys-Lys, instead caused an unwanted increase in liver uptake as well as an accumulation of radioactivity in the kidneys.
  • the inventive compound thus provides the structural modification ma- Xaa1 -Xaa2-Xaa3- on a functional or "effector" biomolecule R, which may be a polypeptide, a DNA molecule, an RNA molecule, a PNA molecule or a mixture of any two or more of these.
  • the structural modification provides a chelating moiety available for binding of a radioactive metal atom M.
  • the biomolecular moiety R provides at least one specific biological function, for example selected from the group consisting of selective interaction with a target substance, in which case R may comprise a targeting moiety for directing the inventive compound towards a specific molecular structure; enzymatic action; therapeutic action; and combinations thereof.
  • the R moiety is capable of selective interaction with a target substance. This may interchangeably be expressed as the R moiety "being capable of binding to" or “having (a binding) affinity for" the target substance, etc.
  • Target substances are preferably structures found inside, on the surface of, or outside cells in the body of a mammalian subject, and are preferably relevant target substances for a medical application, such as cancer diagnostics and/or therapy.
  • Target substances may be molecules bound to a cell surface; molecules bound to intracellular structures; intracellular molecules; components of extracellular matrix; or extracellular molecules.
  • R being capable of selective interaction with a molecule bound to a cell surface, such a molecule may be selected from cell surface antigens, cell surface receptors, cell adhesion molecules and cell surface bound products of a reporter gene within the cell nucleus.
  • Non-limiting examples of molecules bound to cell surfaces are PSMA, EpCAM, CEA, GRPR, NMBR, BRS3, CCK1 R, GASR, NTR-1 , NPY-Y2R, GLP1 R, NK1 R, VPAC1 R, VPAC2R, PACR, EDNR, SSR1 -5, TRLR1 -13, uPAR, PPARs, GLUTR, CD antigens including CD4, CD11 a, CD25, CD44, CD52, CD54, CD56, CD64, CD133, CD135, CD213a, CD213b; tyrosine kinase receptors including EGFR, HER2, ERBB3, ERBB4, cMET, RON, Tie1 , Tie2, IGF1 R, IR, antigens specific for neovasculature including VEGFR1 -3, PDGF receptors ⁇ and ⁇ ; G-protein coupled receptors (GPCR) including chemokine receptors; a
  • R being capable of selective interaction with a molecule bound to an intracellular structure
  • non-limiting examples of such a molecule are cytoskeletal proteins, keratin family, lamins, vimentin, desmin, actin, tubulin, GFAP and melanin.
  • non-limiting examples of such a molecule are kinases, phosphatases and transcription factors, including members of the MAPK kinase, JAK-STAT, EGFR, VEGFR, insulin, integrin, mTOR, NF- ⁇ B, Notch, P53, Wnt, TGFb, TNFRs, ToIIR, FAK and phosphatidylinositol signalling pathways, molecules involved in apoptosis including members of the caspase cascade and those involved in the mitochondrial pathway for apoptosis and cell cycle progression including cyclins and cyclin dependent kinases, as well as protein regulatory molecules including the SOCS box containing proteins, ubiquitinating enzymes, sumoylating proteins, the proteasome complex, heat shock proteins and melanin.
  • non-limiting examples of such a molecule are cytokines, such as members of the TNF and IFN families, interleukins and colony stimulating factors, chemokines including the C, CC, CXC and CX3C families, hormones, such as oestrogen, GH, PRL, erythropoietin, activin, TSH, LSH, ACTH, melatonin, NPY, gastrin, and growth factors, such as FGFs, PDGFs, VEGFs, IGFI s, TGFb, EGF, Heregulin and Neuregulins, plasma proteins, including enzymatically active proteins or inhibitors such as the MMP, ADAM and serpin families, high abundant serum proteins, complement factors, hormone and cytokine binding factors and proteins involved in lipid transportation.
  • cytokines such as members of the TNF and IFN families, interleukins and colony stimulating factors
  • chemokines including the C, CC, CXC and CX3C
  • R being capable of selective interaction with components of extracellular matrix
  • non-limiting examples of such molecules are collagen and laminin families, fibronectin, elastin, endostatins, thrombospondins, tenascin and fibulin.
  • the moiety R having a biological function, may be a polypeptide.
  • R is a polypeptide with from 3 to 200 amino acid residues.
  • R may have from 6 to 120 amino acid residues, for example from 30 to 70 amino acid residues.
  • R may be a polypeptide with from 3 to 30 amino acid residues, for example from 3 to 10 amino acid residues, such as 8 amino acid residues.
  • Such peptides may be comprised in the biological function moiety R in a chelating compound according to the invention, and be labeled with a radionuclide via the ma-Xaa1 -Xaa2-Xaa3 chelating moiety.
  • R may comprise a polypeptide which is selected from peptide targeting agents based on somatostatin, bombesin, CCK, neurotensin, NPY, GLP1 and NK1.
  • R may comprise an antibody molecule, or an antibody fragment or an antibody derivative, which is capable of selective interaction with a target substance (i.e. its antigen).
  • a target substance i.e. its antigen.
  • antibodies, antibody fragments and antibody derivatives are polyclonal antibodies, monospecific antibodies, monoclonal antibodies, Fab' fragments, scFv:s and domain antibodies (dAb:s).
  • R comprises a binding polypeptide, i.e. a polypeptide capable of selective interaction with a target substance, which is derived from a non-antibody scaffold.
  • a binding polypeptide may be engineered using a naturally occurring protein, or domain(s) thereof, as a scaffold (i.e. a starting-point structure).
  • Protein engineering may for example be used to obtain libraries of polypeptide variants, from which libraries suitable binding polypeptides are subsequently selected.
  • Such libraries may be constructed by combinatorial protein engineering, making possible the evolution of highly specific binding polypeptides through randomization of a given number of amino acid residues in the scaffold polypeptide. Suitable procedures for isolation of binding polypeptides from combinatorial libraries are e.g.
  • Non-limiting examples of non-antibody scaffolds for use as starting point for the development of binding polypeptides are three-helix domains, lipocalins, ankyrin repeat domains, cellulose binding domains, Y crystallins, green fluorescent protein, human cytotoxic T lymphocyte-associated antigen 4, protease inhibitors, PDZ domains, peptide aptamers, staphylococcal nuclease, tendamistats, fibronectin type III domain, zinc fingers, avimers, microproteins and conotoxins.
  • R comprises a binding polypeptide which has been engineered using as scaffold a three-helix polypeptide domain.
  • three-helix polypeptide domains are any three- helix domain of Protein G from Streptococcus spp., such as the albumin binding domain (ABD) of Protein G from Streptococcus pyogenes strain G418; any domain of Protein A from Staphylococcus aureus, such as the protein Z variant of the B domain of Protein A from Staphylococcus aureus.
  • R comprises a binding polypeptide which has been engineered using as scaffold a three-helix polypeptide domain with structural properties derived from any protein A domain, including variants displaying alternative surfaces both on the target interaction face and on other sides of the molecule.
  • R comprises a polypeptide with an amino acid sequence selected from those listed below in Table 1.
  • amino acid sequences are given in the standard one-letter code. Table 1 Non-limiting examples of amino acid sequences that may be included in R
  • R in the inventive compound may also comprise a polynucleotide molecule, such as DNA, RNA, PNA or mixtures thereof.
  • a polynucleotide molecule may also be capable of selective interaction with a target substance.
  • aptamers or decoys fold into well-defined three-dimensional structures and may bind to a target substance with high affinity and specificity (see for review Bunka et al, Nat Rev Microbiol 4(8):588-596 (2006)).
  • Hicke et a/ J Nucl Med 47(4):668-678 (2006) reported the use of aptamers that bind to the extracellular matrix protein tenascin-C for imaging of brain and breast tumor xenografts.
  • Another type of polynucleotide targeting agents are polynucleotides that are complementary to the DNA or mRNA of genes amplified or over-expressed in tumors (i.e. antisense polynucleotides).
  • Any polynucleotide ligand for use as the R moiety in the present invention may be of RNA, DNA or PNA origin, or be a mixture of nucleotides with different backbones.
  • the chelating compound according to the invention is suitably produced by known processes.
  • R being a polypeptide
  • the compound may be prepared using expression of a gene encoding the amino acid sequence of interest, optionally including the three amino acid residues of the chelating moiety ma-Xaa1 -Xaa2-Xaa3-.
  • the mercaptoacetyl group may then be conjugated to the purified polypeptide via known coupling chemistry.
  • the compound can be produced by chemical synthesis. By choosing a synthetic production route rather than recombinant expression, the chelating moiety can be introduced site-specifically and the modified peptide can be purified to homogeneity.
  • a chelating compound according to the first aspect of the invention may be complexed with a radionuclide for the purposes of various medical applications.
  • the radioactive metal atom is suitable for in vivo imaging applications, i.e. applications wherein it is of interest to obtain images of the distribution of the radioactive metal atom within a body of a subject.
  • a radionuclide for use in such applications is suitably one that emits gamma radiation with an energy of approximately 100-300 keV or positrons.
  • various isotopes of technetium are preferred metal atoms for use in imaging applications.
  • M is selected from the group consisting of 99m Tc, 94 Tc and 96 Tc, preferably 99m Tc.
  • SPECT Single Photon Emission Computer Tomography
  • PET Positron Emission Tomography are the two techniques that are most frequently used for in vivo detection and visualization of radionuclide distribution in mammalian subjects.
  • SPECT utilizes gamma-quanta, which are emitted from the nuclei of radioactive nuclides.
  • 99m Tc (with a half-life of 6 h) has preferably been used in SPECT, due to the energy of gamma-quanta emitted (140 keV; nearly ideal for gamma-camera/SPECT imaging) and to its short half-life, which considerably reduces the radiation burden on patients.
  • 99m Tc half-life is compatible with imaging using polypeptides.
  • a chelate according to the invention can be used for in vivo imaging.
  • Chelates of the inventive compound with a radionuclide possess physio- chemical properties which in vivo results in improved biodistribution, compared with previously known compounds, such as the previously described 99m Tc:ma-Gly-Gly-Gly based chelates.
  • previously known compounds such as the previously described 99m Tc:ma-Gly-Gly-Gly based chelates.
  • the present inventors have carried out biodistribution studies in mice (normal NMRI mice or Balb/C nu/nu mice bearing SKOV-3 xenografts) and gamma-camera imaging, which visualized the distribution of chelates.
  • the inventive chelate was found to give a lowered hepatobiliary excretion and a shift in excretion to the renal pathway. Chelates of the inventive compound thus reduce the accumulation of radioactivity in the liver and the intestines and makes possible for example tumor imaging with high contrast in the abdomen area at the day of injection. Unexpectedly and importantly, the renal uptake of a chelate of the inventive compound was only slightly increased in comparison with known chelates. Instead, renal retention was low, which is considered to be an effect of prevented re-absorption of the compound from primary urine in proximal tubulae of the kidneys. Reduction of renal retention for such a chelate thus decreases the radioactivity uptake in the kidney.
  • a method of preparing the chelate according to the second aspect comprises mixing, in the presence of a reducing agent, of a compound according to the first aspect of the invention with a radioactive moiety MO 4 " , wherein M is a radioactive metal atom.
  • the initial chemical and isotopic form of the radioactive moiety may be selected from ["" 1 Tc]TcO 4 " , [ 94 Tc]TcO 4 " and [ 96 Tc]TcO 4 " , preferably [ 99m Tc]TcO 4 ⁇
  • mixing is carried out in the presence of a reducing agent.
  • a reducing agent may be SnCI 2 or SnF 2 .
  • the mixing is carried out in the additional presence of a weak chelator, which acts as an intermediate in the transfer of M to the inventive chelating compound.
  • a weak intermediate chelator are tartrate, glucoheptonate and citrate.
  • the step of obtaining an image is repeated at least three times, whereby a series of images is obtained.
  • This embodiment is useful when one seeks to follow the biodisthbution over time of a targeting agent, and allows for pharmacokinetic studies. The skilled person appreciates that any number of images could be obtained, in order to achieve the requisite degree of time resolution in such a study.
  • the method comprises, before the administration step, a preparatory step of preparing a chelate according to the second aspect of the invention, which step comprises mixing, in the presence of a reducing agent, of a compound according to the first aspect with a compound selected from ["" 1 Tc]TcO 4 " , [ 94 Tc]TcO 4 " and [ 96 Tc]TcO 4 " , preferably [ 99m Tc]TcO 4 ⁇
  • a weak intermediate chelator as discussed above may also be used.
  • the present invention provides the chelate according to the second aspect of the invention for use in diagnostics. Also provided is use of said chelate in the preparation of a diagnostic agent for imaging in vivo of the body of a mammalian, including human, subject.
  • Figure 1 illustrates a gamma-camera image of tumor-bearing mice using 99m Tc:ma-Gly-Gly-Gly-Z H ER2342 as targeting agent. Arrows point to caecum (C), kidneys (K) and tumors (T).
  • Figure 2 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Gly-Gly-Gly-Z HE R2342 ( 99m Tc-maGGG-Z HE R2 342 ) and 99m Tc:ma-Ser- Ser-Ser-Z H ER2342 ( 99m Tc-maSSS-Z H ER2342) in normal NMRI mice 4 h post- injection (p.i.).
  • Figure 3 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Ser-Ser-Ser-Z HE R2342 ( 99m Tc-maSSS-Z HE R2342) in Balb/C nu/nu mice bearing HER2-expressing SKOV-3 xenografts 4 h p.i.
  • Figure 4 illustrates a gamma-camera image of a mouse bearing HER2- expressing SKOV-3 xenograft using 99m Tc:ma-Ser-Ser-Ser-Z H ER2342- Arrows point to caecum (C), kidneys (K) and tumors (T).
  • FIG. 5 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Ser-Ser-Ser-Z HE R2 342 ( 99m Tc-maSSS-Z HE R2342), 99m Tc:ma-Glu-Glu- Glu-Z HE R2342 ( 99m Tc-maEEE-Z HE R2342) and 99m Tc:ma-Lys-Lys-Lys-Z HE R2342 ( 99m Tc-maKKK-Z H ER2342) in normal NMRI mice 4 h p.i.
  • FIG. 6 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Ser-Ser-Ser-Z HE R2342 ( 99m Tc-maSSS-Z HE R2342), 99m Tc:ma-Glu-Glu- Glu-Z HER2 342 ( 99m Tc-maEEE-Z HE R2342), 99m Tc:ma-Glu-Glu-Ser-Z HE R 2 342 ( 99m Tc- maEES-Z HE R2342), 99m Tc:ma-Ser-Glu-Glu-Z HER 2342 ( 99m Tc-maSEE-Z HER 2342) and 99m Tc:ma-Glu-Ser-Glu-Z HE R2342 ( 99m Tc-maESE-Z HE R2342) in normal NMRI mice 4 h p.i.
  • Figure 7 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Ser-Ser-Ser-Z HE R2342 ( 99m Tc-maSSS-Z HE R 2 342), 99m Tc:ma-Ser-l_ys- Ser-Z HER 2342 ( 99m Tc-maSKS-Z HE R2342) and 99m Tc:ma-Lys-Lys-Lys-Z HER 2342 ( 99m Tc-maKKK-Z HE R2342) in normal NMRI mice 4 h p.i.
  • FIG. 8 is a diagram showing the biodistribution of targeting agents 99m Tc:ma-Ser-Ser-Ser-Z HE R2342 ( 99m Tc-maSSS-Z HE R 2 342), 99m Tc:ma-Glu-Glu- Glu-Z HER2 342 ( 99m Tc-maEEE-Z HE R2342), 99m Tc:ma-Ser-Glu-Glu-Z HE R 2 342 ( 99m Tc- maSEE-Z HE R 2 342), 99m Tc:ma-Glu-Glu-Ser-Z HE R 2 342 ( 99m Tc-maEES-Z HE R 2 342), and 99m Tc:ma-Glu-Ser-Glu-Z HE R2342 ( 99m Tc-maESE-Z HE R2342) in Balb/C nu/nu mice bearing SKOV-3 xenografts 4 h p.i.
  • Figure 9 is a diagram showing the biodistribution of targeting agent 99m Tc:ma-Ser-Lys-Ser-Z HE R2342 ( 99m Tc-maSKS-Z HE R2342) in Balb/C nu/nu mice bearing SKOV-3 xenografts 4 h p.i.
  • Figure 10 illustrates a gamma-camera image of a mouse bearing HER2-expressing SKOV-3 xenograft using 99m Tc:ma-Glu-Ser-Glu-Z HE R2342- Arrows point to kidneys (K) and tumors (T).
  • Figure 1 1 illustrates the specificity of uptake of 99m Tc:ma-Ser-l_ys-Ser-
  • ZH E R2 342 and 99m Tc:ma-Glu-Ser-Glu-Z HE R2342 in HER2-expressing tumor xenografts were pre-injected in the animals.
  • Data are presented as average percent of injected activity per gram (% IA/g) in four animals ⁇ SEM.
  • tumor cells overexpress the protein kinase HER2 on their surfaces.
  • Polypeptides capable of selective interaction with HER2 have been described, such as a monoclonal antibody to HER2 (trastuzumab, marketed as Herceptin® by Genentech and Roche) and engineered combinatorial proteins, for example those designated
  • the HER2 binding molecule Z HER2 342 was synthesized on a 433A Peptide Synthesizer (Applied Biosystems) using standard Fmoc chemistry, essentially as described in Engfeldt et al, Chembiochem 6:1043-50 (2005).
  • KKLNDAQAPK SEQ ID NO:2
  • Fmoc amide resin with a substitution level of 0.67 mmol/g.
  • the N-terminal Fmoc group was removed by 20 % piperidine-NMP.
  • Acylation reactions were performed in NMP with 10 molar equivalents of amino acid, activated with 2-(1 H-benzothazol-1-yl)- 1 ,1 ,3,3-tetramethyluronium hexafluorophosphate (HBTU) and 1 - hydroxybenzothazole (HOBt) (Advanced ChemTech, Louisville, USA).
  • HBTU 2-(1 H-benzothazol-1-yl)- 1 ,1 ,3,3-tetramethyluronium hexafluorophosphate
  • HBt 1 - hydroxybenzothazole
  • the 17 underlined residues in the sequence above were double coupled. Unreacted amino groups were capped with acetic anhydride (Applied
  • the chelating functions ma-Gly-Gly-Gly-Gly, ma-Ser-Ser-Ser, ma-Glu-Glu- GIu and ma-Lys-Lys-Lys (all for comparison) and ma-Ser-Glu-Glu, ma-Glu- Ser-Glu, ma-Glu-Glu-Ser and ma-Ser-Lys-Ser (illustrating the invention) were introduced by manual synthesis.
  • the N-terminal Fmoc protecting group was removed by incubation with 20 % piperidine-NMP for 20 min.
  • the final deprotection and release of peptides from the resin support was performed as a one-step procedure by treatment with TFA/ethanedithiol(EDT)/thisopropylsilane (TIS)/H 2 O (94:2.5:2.5:1 ) for 2 h. This was followed by three rounds of extraction in te/t-butyl methyl ether-H 2 O (50:50) (Merck), filtration and lyophilization.
  • the product was analyzed by analytical RP-HPLC using a 4.6 x 150 mm polystyrene/divenylbenzene matrix column with a particle size of 5 ⁇ m (GE Healthcare, Sweden), a 20 min gradient of 20-60 % B (A: 0.1 % TFA-H 2 O, B: 0.1 % TFA-CH 3 CN) and a flow rate of 1 ml/min.
  • the peptides were purified using a 20 min gradient of 25-45 % B.
  • the pure product was lyophilized prior to dissolution in 10 mM NH 4 Ac pH 5,5, and the final peptide concentration was determined by amino acid analysis.
  • ZHER2342 was successfully synthesized using Fmoc solid phase peptide synthesis.
  • the synthetic yield was determined to 21 % by analytical RP- HPLC.
  • the total yield of chelating polypeptides was 13-17 %.
  • the peptides were purified to more than 90 % purity.
  • the mass of each peptide was determined by ESI-MS, and the experimentally determined molecular weights correlated well with the theoretically calculated values (Table 2).
  • Variable temperature measurements were performed using a Jasco J- 810 spectropolahmeter (JASCO, Tokyo, Japan). Samples were diluted to a concentration of 50 ⁇ M in PBS, pH 7.4. A cell with an optical path length of 1 mm was used. CD spectra from 250 to 195 nm were obtained at 20 0 C before and after melting tests. For melting point measurements, the absorbance was measured at 221 nm and the temperature was increased by 5 °C/min from 20 to 90 0 C.
  • HBS HEPES
  • NaCI sodium EDTA
  • 0.005 % Surfactant P20 pH 7.4
  • the flow rate was set to 50 ⁇ l/min with HBS as running buffer, and the samples were injected for 5 min followed by 10 min dissociation and regeneration with 20 ⁇ l HCI (20 mM).
  • the BiaEval software Biacore
  • the Langmuir 1 :1 binding model were used for the kinetic analysis.
  • the 99m Tc-labeled Z HER2 342 variants were isolated from unreacted technetium and other low-molecular weight components using size-exclusion chromatography with a disposable NAP-5 column (GE Healthcare, Sweden) pre-equilibrated with PBS. Separation was performed according to the manufacturer's instruction.
  • PBS as an eluent made it possible to obtain 99m Tc-labeled Z HER2 342 directly in an injectable form after purification. Losses of labeled conjugate during purification were on the order of 13-17 %. According to ITLC analysis of the purified product, its purity was always higher than 95 %. Labels were stable in PBS.
  • Stability tests were performed, in which labeled Z HER2 342 molecules were incubated in PBS or in PBS with 300 molar equivalents of cysteine. Stability was analyzed by ITLC. After a one hour incubation with a 300-fold excess of cysteine at 37 0 C, more than 90 % of the radioactivity was associated with the Z HER2 342 molecule, which indicates strong binding of 99m Tc. Binding specificity of " m Tc-labeled Z H ER2342 variants to HER2-expressing SKOV-3 cells
  • SKOV-3 cells bearing 1.2 x 10 6 HER2 receptors per cell were cultivated on Petri dishes with a diameter of 3.5 cm to a cell density of 2-5 x 10 5 CeIIs per dish.
  • Labeled conjugates 0.8 ng/10 5 cells, were added to two groups of Petri dishes containing SKOV-3 cells. One group of dishes was pre-saturated with a 1000-fold excess of non-labeled recombinant Z HER2 342 molecules 10 min before labeled molecules were added. The cells were incubated for 1 h at 37 0 C and incubation media was collected. The cell dishes were washed with cold serum-free medium and treated with 0.5 ml trypsin-EDTA solution (0.05 % trypsin, 0.02 % EDTA in PBS buffer, Flow Irvine, UK) for 10 min at 37 0 C. When cells were detached, 0.5 ml complete medium was added to every dish and cells were re-suspended.
  • trypsin-EDTA solution 0.05 % trypsin, 0.02 % EDTA in PBS buffer, Flow Irvine, UK
  • the cell suspension was collected for radioactivity measurements.
  • Cell-associated radioactivity (C) was measured on an automated gamma-counter with a 3-inch NaI(TI) detector (1480 WIZARD, Wallac OY, Finland) in parallel with 1 ml corresponding incubation medium (M).
  • M incubation medium
  • the results of this binding specificity experiment showed that binding of all 99m Tc-labeled Z HER2 342 chelators could be prevented by receptor saturation (data not shown).
  • mice Female outbred Balb/c nu/nu mice (10-12 weeks old at arrival) were used in the in vivo experiments.
  • SKOV-3 tumors were grafted by subcutaneous injection of approximately 7 x 10 6 cells in the right hind leg. Xenografts were allowed to develop during eight weeks. Biodistribution in normal NMRI mice
  • mice were injected with approximately 1 ⁇ g 99m Tc-labeled Z H ER2342 (approximately 100 kBq). The biodistribution measurement was performed 4 h after injection (pi). Mice were intraperitoneal ⁇ injected with a lethal dose of ketamine HCI
  • mice were killed with heart puncture with a 1 ml syringe rinsed with diluted heparin (Leo Pharma).
  • Organs and tissue samples of blood, lung, liver, spleen, kidney, stomach, salivary glands, thyroid and muscle were excised and collected in weighed plastic bottles. Intestines were collected together with their content. Organs and tissue samples were weighed and their radioactivity was recorded using an automatic gamma-spectrometer with standard 99m Tc protocol. The background was measured for each sample holder in the same way. Tissue uptake values were calculated as percent of injected activity per gram tissue (% IA/g).
  • Radioactivity in kidneys and intestine with contents were selected as a measure of renal and hepatobiliary excretion. Other organs were selected to evaluate in vivo stability of the technetium label.
  • An elevated level of blood radioactivity would be an indication of transchelation of 99m Tc to blood plasma proteins, while formation of radio colloids or macroaggregates should be reflected in an elevated uptake in liver, spleen or lungs. Re-oxidation and release of pertechnetate during catabolism of the label should cause increased uptake of radioactivity in stomach, salivary gland and thyroid.
  • mice bearing SKOV-3 xenografts were used.
  • 99m Tc:ma-Glu-Ser-Glu- ZHER2342 and 99m Tc:ma- Ser-Lys-Ser-Z H ER2342 an extra group of four mice was subcutaneously injected with 750 ⁇ g of non-labeled recombinant Z HER2 342 in 250 ⁇ l PBS 1 h before the radioactive injection (blocking experiment).
  • mice were subcutaneously injected with the various 99m Tc:ma- Xaa1 -Xaa2-Xaa3-Z H ER2342 variants in 100 ⁇ l PBS (approximately 100 kBq).
  • mice were injected intraperitoneal ⁇ with a lethal dose of ketamine HCI (Ketalar®, Pfizer) and xylazine HCI (Rompun®, Bayer).
  • the mice were killed with heart puncture with a 1 ml syringe rinsed with diluted heparin (Leo Pharma).
  • the mice in the blocking group were killed 4 h pi.
  • the samples were treated as in the previously described experiments on biodistribution in normal NMRI mice (see above).
  • mice bearing SKOV-3 xenografts were intravenously injected (tail) with 3 MBq (3 ⁇ g) 99m Tc:ma-Gly-Gly-Gly-Z H ER2342 (comparison), 9 9m Tc:ma-Ser-Ser-Z H ER2342 (comparison) or 99m Tc:ma-Glu-Ser-Glu- Z HER2 342 (illustrating the invention).
  • the biodistribution experiment in normal mice demonstrated that the use of the ma-Ser-Ser-Ser chelator enabled a reduction of the level of hepatobiliary excretion three-fold, at a cost of moderate increase of radioactivity uptake in the kidneys ( Figure 2).
  • the biodistribution experiment in tumor-bearing mice confirmed the capacity of 99m Tc:ma-Ser-Ser-Ser-Z H ER2342 to target HER2-expressing tumor xenografts. Radioactivity accumulation in the tumors exceeded the accumulation in all organs and tissues, except the kidneys ( Figure 3). The tumor accumulation was even higher than the average accumulation in the gastrointestinal tract, with account taken of the fact that the typical sample weight of intestines was 3 gram.
  • a gamma camera image ( Figure 4) demonstrated improvement in contrast in comparison with 99m Tc:ma-Gly-Gly-Gly-Z H ER2342- Still, a radioactivity accumulation in caecum was clearly seen, which caused a modest contrast between tumor and abdominal area.
  • Kidney accumulation was 95 ⁇ 23 % IA/g (percent injected activity per gram) for 99m Tc:ma-Glu-Glu-Glu-Z H ER2342 and 127 ⁇ 9 % IA/g for 99m Tc:ma-Lys-Lys-Lys-Z H ER2342 (compared with 18 ⁇ 4 % IA/g for 99m Tc:ma-Ser-Ser-Ser-Z H ER2342)
  • use of ma-Lys-Lys-Lys as a chelator caused high radioactivity uptake in liver, 6.8 ⁇ 2.5 % IA/g (compared with 0.48 ⁇ 0.28 % IA/g for 99m Tc:ma-Ser-Ser-Z HE R2 342 and 0.21 ⁇ 0.01 % IA/g for 99m Tc:ma-Glu-Glu-Z HE R2342).
  • the tumor-targeting properties of 99m Tc:ma-Glu-Glu-Glu-Z H ER2342, 99m Tc:ma-Ser-Glu-Glu-Z HE R2342, 99m Tc:ma-Glu-Glu-Ser-Z HE R2342, 99m Tc:ma- Glu-Ser-Glu -Z H ER2342, and 99m Tc:ma-Ser-Lys-Ser-Z H ER2342 were verified in mice bearing HER2-expressing SKOV-3 xenografts ( Figures 8 and 9).
  • the biodistribution of the inventive chelating compounds was characterized by quick clearance of radioactivity from blood and all organs and tissues.
  • the radioactivity concentration in tumors was higher than in other organs or tissues except for the kidneys.
  • gamma-camera imaging of 99m Tc:ma-Glu-Ser-Glu-Z H ER2342 ( Figure 10) showed that high contrast images of HER2-expressing xenografts in mice can be produced using a conjugate according to the invention.
  • the only organ with apparent accumulation of radioactivity apart from the tumor was the kidney.
  • kidneys in the body are well-defined, which would help in the interpretation of the gamma-camera images.
  • the reduction in kidney uptake further reduces this problem.
  • radioactivity in the abdominal area was very low, and a high contrast of tumor imaging in this area was possible.

Abstract

A chelating compound of the formula ma-Xaa1-Xaa2-Xaa3-R is provided, wherein - ma is mercaptoacetyl; - Xaa1, Xaa2 and Xaa3 are amino acid residues defined as follows: o any one or two of Xaa1, Xaa2 and Xaa3 is/are the amino acid residue Ser; o in case two of Xaa1, Xaa2 and Xaa3 are Ser, the remaining amino acid residue is selected from the group consisting of Glu, Arg and Lys; o in case one of Xaa1, Xaa2 and Xaa3 is Ser, the remaining amino acid residues are both Glu; and - R has a biological function and is selected from the group consisting of polypeptides, PNA, DNA, RNA, and mixtures thereof. Also provided are chelates of this compound with radioactive metal atoms, as well as methods and uses employing the chelating compound.

Description

NEW CHELATING COMPOUND
Field of the invention
The present invention relates to compounds useful for in vivo imaging. More particularly, the invention relates to a chelating compound and a chelate thereof with a radioactive metal atom. The present invention furthermore relates to methods of diagnosis, wherein the chelating compound is used.
Background of the invention
Molecular targeting of chemical structures within the body of a subject has been evaluated, for example for purposes of in vivo diagnostics of cancer disease. The targeting of tumor specific chemical structures is called tumor targeting. Molecules used for detection in the various applications of molecular targeting are called targeting agents. Monoclonal antibodies, fragments of monoclonal antibodies and peptides have been evaluated as targeting agents. The large size of antibodies, however, causes slow blood clearance, slow extravasation and slow tumor penetration. Therefore, the use of peptide receptor ligands for targeting of receptors that are overexpressed in malignant tumors is today a common approach in radionuclide tumor targeting. The small size of such peptides improves extravasation and tumor penetration. At the same time, a large proportion of short peptides (smaller than 60 kD) is rapidly cleared via the kidneys. This reduces radioactivity concentration in blood and healthy tissues. In this way, a high contrast (tumor-to-non-tumor ratio) is obtained when the peptides are used in the context of in vivo imaging. However, peptides are often re-absorbed in proximal tubulae after glomerular filtration, which can cause the accumulation of excessive radioactivity in the kidneys. Such high radioactivity accumulation in kidneys complicates the imaging of targets located close to the kidneys.
US 4 861 869 discloses bifunctional coupling agents suitable for forming conjugates with biomolecules and for complexing with radionuclide atoms. Examples of such coupling agents are various salts of modified mercaptoacetyl-glycyl-glycyl-glycinate, which are coupled to different antibody variants.
EP 284 071 A2 discloses similar labeling of biomolecules with a mercaptoacetyl-glycyl-glycyl-glycyl moiety for complexing with a radionuclide. US 5 980 861 is directed to the labeling of polynucleotides via a chelator compound, which chelator compound is either mercaptoacetyl-glycyl- glycyl-glycyl or mercaptoacetyl-seryl-seryl-seryl.
The above references are examples of using a tetradentate chelating group, which comprises three amide nitrogens from a peptide backbone and a sulfur from the terminal mercapto group. Formula (II) on page 6 of
EP 284 071 A2 is a schematic illustration of the geometry of such a complex.
Chelating groups such as those described in the above references are collectively denoted N3S or N3S chelators, due to the three N's and one S involved. The work to date relating to such N3S chelators has been centered on mercaptoacetyl-glycyl-glycyl-glycyl, which in the present text is denoted ma-Gly-Gly-Gly. It is also referred to in the literature as MAG3 or MAG3. ma-Gly-Gly-Gly with different protective groups has been utilized for labeling of antibodies (Lei et al, Nucl Med Biol 23(7):917-922 (1996)) and their
Fab (Kim et al, Nucl Med Biol 29(2):139-142 (2002)) and dsFv fragments (Kobayashi et al, Cancer Res 56:3788-3795 (1996)), peptides (Qu et al, Nucl
Med Commun 22(2):203-215 (2001 ); Dechstoforo and Mather, Bioconjugate
Chem 10:431 -438 (1999)), and antisense DNA (Zhang et al, Eur J Nucl Med
27(11 ):1700-1707 (2000)), demonstrating good in vivo stability and, often, good pharmacokinetics. Some work has also been done on alternatives to ma-Gly-Gly-Gly.
Thus, in Appl Radiat lsot 50(4):723-732 (1999), Chang et al report on studies performed on mercaptoacetyl-seryl-seryl-seryl, i.e. ma-Ser-Ser-Ser, also denoted MAS3 or MAS3.
Other alternative N3S chelators were studied by Zhu et al in Nucl Med Biol 28:703-708 (2001 ). In order to develop and test an alternative synthesis route, the authors synthesized ma-Gly-Gly-Gly as well as glutamic acid or phenylalanine containing variants ma-Glu-Gly-Gly and ma-Phe-Gly-Gly. The effect on biodistribution in mice was studied via coupling of these chelators to the 7 kD HNE2 protein.
Another biodistribution study was carried out by Verbeke et al and reported in Nucl Med Biol 27:769-779 (2000). However, this group used another type of chelator with only two amino acid residues coupled to the mercaptoacetyl group.
Previous experiments with mercaptoacetyl-thpeptide chelators, N3S chelators, have not yielded optimal results with regard to excretion pathways of the radiolabeled targeting agent. In particular, excretion via the hepatobiliary pathway leads to significant accumulation of activity in the liver and the intestines. This may necessitate imaging the day after injection, instead of imaging at the day of injection, in order to detect expression in extrahepatic metastases in the abdomen area and ovarian carcinoma. Therefore, it is desirable to effect a shift from the hepatobiliary pathway to the renal pathway. At the same time, when the desired properties of reduced hepatobiliary excretion are balanced by a high rate of clearance by the kidneys, it is desirable to minimize re-absorption in the kidneys. No chelator moiety for conjugation with a targeting agent has hitherto been presented which offers the requisite balance in this regard. Therefore, improved targeting agents are called for, which reduce the radioactivity concentration in healthy organs and at the same time enable high sensitivity in for example in vivo imaging.
Description of the invention One object of the present invention is to meet this demand, by developing new and improved chelating compounds that can function as targeting agents when complexed to a suitable radioactive metal atom.
Another object of the present invention is to provide chelates of such compounds with radioactive metal atoms, which chelates show high sensitivity for target structures.
A related object is to provide chelates of such compounds with radioactive metal atoms, which chelates reduce the radioactivity accumulation in healthy tissue. Yet another object of the present invention is to provide methods for preparing said chelates of compounds with radioactive metal atoms.
Still another object of the present invention is to provide means and methods that enable targeted in vivo imaging for diagnostic and/or pharmacokinetic studies.
Thus, in a first aspect of the invention, there is provided a chelating compound of the formula: ma-Xaa1 -Xaa2-Xaa3-R wherein - ma is mercaptoacetyl;
- Xaa1 , Xaa2 and Xaa3 are amino acid residues defined as follows: o any one or two of Xaa1 , Xaa2 and Xaa3 is/are the amino acid residue Ser; o in case two of Xaa1 , Xaa2 and Xaa3 are Ser, the remaining amino acid residue is selected from the group of amino acid residues consisting of GIu, Arg and Lys; o in case one of Xaa1 , Xaa2 and Xaa3 is Ser, the remaining amino acid residues are both GIu residues.
- R has a biological function and is selected from the group consisting of polypeptides, PNA, DNA, RNA, and mixtures thereof.
Thus, the chelating compound according to the invention has a chelating moiety, ma-Xaa1 -Xaa2-Xaa3, and a moiety conferring a biological function, R. In the present specification, variants of the chelating moiety of the inventive chelating compounds are denoted ma-Xaa1 -Xaa2-Xaa3, each of said Xaa:s being an amino acid represented in the standard three-letter code. A chelating moiety in a chelating compound according to the invention, in which moiety Xaa1 is GIu, Xaa2 is Ser and Xaa3 is GIu, is thus denoted ma- Glu-Ser-Glu, GIu and Ser being the three letter representations of glutamic acid and serine, respectively. In one embodiment of the present invention, the ma-Xaa1 -Xaa2-Xaa3 chelating moiety comprises one or two Ser, the remaining two or one amino acid residues being GIu. For example, Xaa3 may be GIu, and the inventive compound be selected from ma-Ser-Glu-Glu-R, ma-Glu-Ser-Glu-R and ma- Ser-Ser-Glu-R. Alternatively, Xaa3 may be Ser, and the inventive compound be selected from ma-Ser-Glu-Ser-R, ma-Glu-Glu-Ser-R and ma-Glu-Ser-Ser- R.
In the experiments described below, a combination of one serine and two glutamic acid residues (ma-Glu-Ser-Glu, ma-Ser-Glu-Glu and ma-Glu- Glu-Ser) was found to significantly reduce renal uptake in comparison with a chelating moiety studied for comparative purposes, ma-Glu-Glu-Glu, in an unexpected fashion. In addition, it was found that accumulated radioactivity in the gastrointestinal tract for the inventive compounds was less than 5 % of the total radioactivity in Balb/C mice, meaning that radiolabeled compounds of the above general formula are useful for imaging of tumors in the abdominal area. Thus, the inventive compounds have good tumor absorption as well as rapid clearance from normal organs and tissue. In imaging applications, radiolabeled targeting agents must have high tumor-to-background ratios, in order to obtain images with a high sensitivity.
In another embodiment, ma-Xaa1 -Xaa2-Xaa3 comprises two Ser, the remaining amino acid residue being selected from Arg and Lys. For example, the remaining amino acid residue may be Arg, and the inventive compound be selected from ma-Ser-Ser-Arg-R, ma-Ser-Arg-Ser-R and ma-Arg-Ser-Ser- R. Alternatively, the remaining amino acid residue may be Lys, and the inventive compound be selected from ma-Ser-Ser-Lys-R, ma-Ser-Lys-Ser-R and ma-Lys-Ser-Ser-R. In a preferred embodiment, the inventive compound is selected from ma-Ser-Arg-Ser-R and ma-Ser-Lys-Ser-R. As demonstrated in the experiments recounted below, the incorporation of one positively charged amino acid residue in the chelating moiety of the inventive compound unexpectedly provides the desired shift in excretion from the hepatobiliary pathway to the renal pathway. At the same time, renal uptake increased only marginally. In a comparative experiment also reported below, a chelating moiety having three positively charged amino acids, e.g. ma-Lys-Lys-Lys, instead caused an unwanted increase in liver uptake as well as an accumulation of radioactivity in the kidneys.
The inventive compound thus provides the structural modification ma- Xaa1 -Xaa2-Xaa3- on a functional or "effector" biomolecule R, which may be a polypeptide, a DNA molecule, an RNA molecule, a PNA molecule or a mixture of any two or more of these. The structural modification provides a chelating moiety available for binding of a radioactive metal atom M.
The biomolecular moiety R provides at least one specific biological function, for example selected from the group consisting of selective interaction with a target substance, in which case R may comprise a targeting moiety for directing the inventive compound towards a specific molecular structure; enzymatic action; therapeutic action; and combinations thereof. In one embodiment, the R moiety is capable of selective interaction with a target substance. This may interchangeably be expressed as the R moiety "being capable of binding to" or "having (a binding) affinity for" the target substance, etc.
Target substances are preferably structures found inside, on the surface of, or outside cells in the body of a mammalian subject, and are preferably relevant target substances for a medical application, such as cancer diagnostics and/or therapy. Target substances may be molecules bound to a cell surface; molecules bound to intracellular structures; intracellular molecules; components of extracellular matrix; or extracellular molecules. In the case of R being capable of selective interaction with a molecule bound to a cell surface, such a molecule may be selected from cell surface antigens, cell surface receptors, cell adhesion molecules and cell surface bound products of a reporter gene within the cell nucleus. Non-limiting examples of molecules bound to cell surfaces are PSMA, EpCAM, CEA, GRPR, NMBR, BRS3, CCK1 R, GASR, NTR-1 , NPY-Y2R, GLP1 R, NK1 R, VPAC1 R, VPAC2R, PACR, EDNR, SSR1 -5, TRLR1 -13, uPAR, PPARs, GLUTR, CD antigens including CD4, CD11 a, CD25, CD44, CD52, CD54, CD56, CD64, CD133, CD135, CD213a, CD213b; tyrosine kinase receptors including EGFR, HER2, ERBB3, ERBB4, cMET, RON, Tie1 , Tie2, IGF1 R, IR, antigens specific for neovasculature including VEGFR1 -3, PDGF receptors α and β; G-protein coupled receptors (GPCR) including chemokine receptors; apoptosis related receptors such as TRAILR 1-4, FASR, receptors within the TNFR family; cell adhesion molecules including integrins, cadherins, selectins, desmosomal cadherins, membrane proteoglycans, desmin, syndecan and ion channels.
In the case of R being capable of selective interaction with a molecule bound to an intracellular structure, non-limiting examples of such a molecule are cytoskeletal proteins, keratin family, lamins, vimentin, desmin, actin, tubulin, GFAP and melanin.
In the case of R being capable of selective interaction with an intracellular molecule, non-limiting examples of such a molecule are kinases, phosphatases and transcription factors, including members of the MAPK kinase, JAK-STAT, EGFR, VEGFR, insulin, integrin, mTOR, NF-κB, Notch, P53, Wnt, TGFb, TNFRs, ToIIR, FAK and phosphatidylinositol signalling pathways, molecules involved in apoptosis including members of the caspase cascade and those involved in the mitochondrial pathway for apoptosis and cell cycle progression including cyclins and cyclin dependent kinases, as well as protein regulatory molecules including the SOCS box containing proteins, ubiquitinating enzymes, sumoylating proteins, the proteasome complex, heat shock proteins and melanin.
In the case of R being capable of selective interaction with an extracellular molecule, non-limiting examples of such a molecule are cytokines, such as members of the TNF and IFN families, interleukins and colony stimulating factors, chemokines including the C, CC, CXC and CX3C families, hormones, such as oestrogen, GH, PRL, erythropoietin, activin, TSH, LSH, ACTH, melatonin, NPY, gastrin, and growth factors, such as FGFs, PDGFs, VEGFs, IGFI s, TGFb, EGF, Heregulin and Neuregulins, plasma proteins, including enzymatically active proteins or inhibitors such as the MMP, ADAM and serpin families, high abundant serum proteins, complement factors, hormone and cytokine binding factors and proteins involved in lipid transportation.
In the case of R being capable of selective interaction with components of extracellular matrix, non-limiting examples of such molecules are collagen and laminin families, fibronectin, elastin, endostatins, thrombospondins, tenascin and fibulin. As stated above, the moiety R, having a biological function, may be a polypeptide. In one embodiment of the invention, R is a polypeptide with from 3 to 200 amino acid residues. In a preferred embodiment, R may have from 6 to 120 amino acid residues, for example from 30 to 70 amino acid residues. In another preferred embodiment, R may be a polypeptide with from 3 to 30 amino acid residues, for example from 3 to 10 amino acid residues, such as 8 amino acid residues.
As described briefly in the Background section, much work has been done developing peptides as targeting agents, targeted towards their peptide receptors (reviewed in Reubi, Endocr Rev 24(4):389-427 (2003)). Such peptides may be comprised in the biological function moiety R in a chelating compound according to the invention, and be labeled with a radionuclide via the ma-Xaa1 -Xaa2-Xaa3 chelating moiety. As a non-limiting list of examples, R may comprise a polypeptide which is selected from peptide targeting agents based on somatostatin, bombesin, CCK, neurotensin, NPY, GLP1 and NK1.
In one embodiment, R may comprise an antibody molecule, or an antibody fragment or an antibody derivative, which is capable of selective interaction with a target substance (i.e. its antigen). Non-limiting examples of antibodies, antibody fragments and antibody derivatives are polyclonal antibodies, monospecific antibodies, monoclonal antibodies, Fab' fragments, scFv:s and domain antibodies (dAb:s).
In another embodiment of the present invention, R comprises a binding polypeptide, i.e. a polypeptide capable of selective interaction with a target substance, which is derived from a non-antibody scaffold. Such a binding polypeptide may be engineered using a naturally occurring protein, or domain(s) thereof, as a scaffold (i.e. a starting-point structure). Protein engineering may for example be used to obtain libraries of polypeptide variants, from which libraries suitable binding polypeptides are subsequently selected. Such libraries may be constructed by combinatorial protein engineering, making possible the evolution of highly specific binding polypeptides through randomization of a given number of amino acid residues in the scaffold polypeptide. Suitable procedures for isolation of binding polypeptides from combinatorial libraries are e.g. phage display, ribosome display, yeast two-hybrid system, mRNA display, SELEX (System Evolution of Ligands by Exponential Enrichment) and protein fragment complementation assays (PCA). Non-limiting examples of non-antibody scaffolds for use as starting point for the development of binding polypeptides are three-helix domains, lipocalins, ankyrin repeat domains, cellulose binding domains, Y crystallins, green fluorescent protein, human cytotoxic T lymphocyte-associated antigen 4, protease inhibitors, PDZ domains, peptide aptamers, staphylococcal nuclease, tendamistats, fibronectin type III domain, zinc fingers, avimers, microproteins and conotoxins.
In a preferred embodiment, R comprises a binding polypeptide which has been engineered using as scaffold a three-helix polypeptide domain. Non-limiting examples of such three-helix polypeptide domains are any three- helix domain of Protein G from Streptococcus spp., such as the albumin binding domain (ABD) of Protein G from Streptococcus pyogenes strain G418; any domain of Protein A from Staphylococcus aureus, such as the protein Z variant of the B domain of Protein A from Staphylococcus aureus. In some embodiments of the invention, R comprises a binding polypeptide which has been engineered using as scaffold a three-helix polypeptide domain with structural properties derived from any protein A domain, including variants displaying alternative surfaces both on the target interaction face and on other sides of the molecule. In specific embodiments of the present invention, R comprises a polypeptide with an amino acid sequence selected from those listed below in Table 1. In Table 1 , amino acid sequences are given in the standard one-letter code. Table 1 Non-limiting examples of amino acid sequences that may be included in R
Figure imgf000011_0001
As an alternative to, or in combination with, the polypeptides discussed above, R in the inventive compound may also comprise a polynucleotide molecule, such as DNA, RNA, PNA or mixtures thereof. Such a polynucleotide molecule may also be capable of selective interaction with a target substance. For example, single stranded nucleic acids, called aptamers or decoys, fold into well-defined three-dimensional structures and may bind to a target substance with high affinity and specificity (see for review Bunka et al, Nat Rev Microbiol 4(8):588-596 (2006)). For example, Hicke et a/ (J Nucl Med 47(4):668-678 (2006)) reported the use of aptamers that bind to the extracellular matrix protein tenascin-C for imaging of brain and breast tumor xenografts. Another type of polynucleotide targeting agents are polynucleotides that are complementary to the DNA or mRNA of genes amplified or over-expressed in tumors (i.e. antisense polynucleotides). Any polynucleotide ligand for use as the R moiety in the present invention may be of RNA, DNA or PNA origin, or be a mixture of nucleotides with different backbones. An example of labeling of a PNA molecule with a known N3S chelator was presented by Chang et al (1999, supra), whereas Zhang et al (J Nucl Med 46(6):1052-1058 (2005)) used a known N3S chelator to label antisense DNA. Analogous procedures may be followed using the novel chelator moiety described herein.
The chelating compound according to the invention is suitably produced by known processes. In the case of R being a polypeptide, the compound may be prepared using expression of a gene encoding the amino acid sequence of interest, optionally including the three amino acid residues of the chelating moiety ma-Xaa1 -Xaa2-Xaa3-. The mercaptoacetyl group may then be conjugated to the purified polypeptide via known coupling chemistry. Alternatively, the compound can be produced by chemical synthesis. By choosing a synthetic production route rather than recombinant expression, the chelating moiety can be introduced site-specifically and the modified peptide can be purified to homogeneity.
As explained in detail above, and in keeping with the various objects of the invention, a chelating compound according to the first aspect of the invention may be complexed with a radionuclide for the purposes of various medical applications. Thus, in a second aspect thereof, the present invention provides a chelate of a chelating compound as described above with a radioactive moiety selected from M, M=O and O=M=O, wherein M is a radioactive metal atom.
In one embodiment of this aspect of the invention, the radioactive metal atom is suitable for in vivo imaging applications, i.e. applications wherein it is of interest to obtain images of the distribution of the radioactive metal atom within a body of a subject. A radionuclide for use in such applications is suitably one that emits gamma radiation with an energy of approximately 100-300 keV or positrons. As is known to the person of skill in the field, various isotopes of technetium are preferred metal atoms for use in imaging applications. Thus, in one embodiment of the present invention, M is selected from the group consisting of 99mTc, 94Tc and 96Tc, preferably 99mTc. SPECT, Single Photon Emission Computer Tomography, and PET, Positron Emission Tomography are the two techniques that are most frequently used for in vivo detection and visualization of radionuclide distribution in mammalian subjects. SPECT utilizes gamma-quanta, which are emitted from the nuclei of radioactive nuclides. 99mTc (with a half-life of 6 h) has preferably been used in SPECT, due to the energy of gamma-quanta emitted (140 keV; nearly ideal for gamma-camera/SPECT imaging) and to its short half-life, which considerably reduces the radiation burden on patients. Furthermore, 99mTc half-life is compatible with imaging using polypeptides.
A chelate according to the invention can be used for in vivo imaging. Chelates of the inventive compound with a radionuclide possess physio- chemical properties which in vivo results in improved biodistribution, compared with previously known compounds, such as the previously described 99mTc:ma-Gly-Gly-Gly based chelates. As will be explained in detail in the Examples, the present inventors have carried out biodistribution studies in mice (normal NMRI mice or Balb/C nu/nu mice bearing SKOV-3 xenografts) and gamma-camera imaging, which visualized the distribution of chelates. The inventive chelate was found to give a lowered hepatobiliary excretion and a shift in excretion to the renal pathway. Chelates of the inventive compound thus reduce the accumulation of radioactivity in the liver and the intestines and makes possible for example tumor imaging with high contrast in the abdomen area at the day of injection. Unexpectedly and importantly, the renal uptake of a chelate of the inventive compound was only slightly increased in comparison with known chelates. Instead, renal retention was low, which is considered to be an effect of prevented re-absorption of the compound from primary urine in proximal tubulae of the kidneys. Reduction of renal retention for such a chelate thus decreases the radioactivity uptake in the kidney. This significant combination, displayed by the present invention, of reduction of hepatobiliary excretion on the one hand and reduction of renal retention on the other, provides the excellent biodisthbution properties needed in many medical applications. In a further, third, aspect of the invention, there is provided a method of preparing the chelate according to the second aspect. The method comprises mixing, in the presence of a reducing agent, of a compound according to the first aspect of the invention with a radioactive moiety MO4 ", wherein M is a radioactive metal atom. In order to prepare a chelate suitable for in vivo imaging applications, the initial chemical and isotopic form of the radioactive moiety may be selected from [""1Tc]TcO4 ", [94Tc]TcO4 " and [96Tc]TcO4 ", preferably [99mTc]TcO4\
In the method according to this aspect of the invention, mixing is carried out in the presence of a reducing agent. As non-limiting examples, said reducing agent may be SnCI2 or SnF2.
Optionally, the mixing is carried out in the additional presence of a weak chelator, which acts as an intermediate in the transfer of M to the inventive chelating compound. Non-limiting examples of such a weak intermediate chelator are tartrate, glucoheptonate and citrate. According to a fourth aspect thereof, the present invention provides a method of in vivo imaging of the body of a mammalian, including human, subject, which method comprises the steps of:
- administering a chelate according to the invention into the body of a mammalian subject; and - obtaining an image of at least a part of the subject's body using a medical imaging instrument, said image indicating the presence of M inside said body. In one embodiment of this inventive method, the step of obtaining an image is repeated at least three times, whereby a series of images is obtained. This embodiment is useful when one seeks to follow the biodisthbution over time of a targeting agent, and allows for pharmacokinetic studies. The skilled person appreciates that any number of images could be obtained, in order to achieve the requisite degree of time resolution in such a study.
In one embodiment of the imaging method according to the invention, the method comprises, before the administration step, a preparatory step of preparing a chelate according to the second aspect of the invention, which step comprises mixing, in the presence of a reducing agent, of a compound according to the first aspect with a compound selected from [""1Tc]TcO4 ", [94Tc]TcO4 " and [96Tc]TcO4 ", preferably [99mTc]TcO4\ Optionally, a weak intermediate chelator as discussed above may also be used. In related aspects, the present invention provides the chelate according to the second aspect of the invention for use in diagnostics. Also provided is use of said chelate in the preparation of a diagnostic agent for imaging in vivo of the body of a mammalian, including human, subject.
The invention will now be illustrated in a non-limiting manner by the following Example and Figures.
Brief description of the figures
Figure 1 illustrates a gamma-camera image of tumor-bearing mice using 99mTc:ma-Gly-Gly-Gly-ZHER2342 as targeting agent. Arrows point to caecum (C), kidneys (K) and tumors (T).
Figure 2 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Gly-Gly-Gly-ZHER2342 (99mTc-maGGG-ZHER2342) and 99mTc:ma-Ser- Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342) in normal NMRI mice 4 h post- injection (p.i.).
Figure 3 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Ser-Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342) in Balb/C nu/nu mice bearing HER2-expressing SKOV-3 xenografts 4 h p.i. Figure 4 illustrates a gamma-camera image of a mouse bearing HER2- expressing SKOV-3 xenograft using 99mTc:ma-Ser-Ser-Ser-ZHER2342- Arrows point to caecum (C), kidneys (K) and tumors (T).
Figure 5 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Ser-Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342), 99mTc:ma-Glu-Glu- Glu-ZHER2342 (99mTc-maEEE-ZHER2342) and 99mTc:ma-Lys-Lys-Lys-ZHER2342 (99mTc-maKKK-ZHER2342) in normal NMRI mice 4 h p.i.
Figure 6 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Ser-Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342), 99mTc:ma-Glu-Glu- Glu-ZHER2342 (99mTc-maEEE-ZHER2342), 99mTc:ma-Glu-Glu-Ser-ZHER2342 (99mTc- maEES-ZHER2342), 99mTc:ma-Ser-Glu-Glu-ZHER2342 (99mTc-maSEE-ZHER2342) and 99mTc:ma-Glu-Ser-Glu-ZHER2342 (99mTc-maESE-ZHER2342) in normal NMRI mice 4 h p.i.
Figure 7 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Ser-Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342), 99mTc:ma-Ser-l_ys- Ser-ZHER2342 (99mTc-maSKS-ZHER2342) and 99mTc:ma-Lys-Lys-Lys-ZHER2342 (99mTc-maKKK-ZHER2342) in normal NMRI mice 4 h p.i.
Figure 8 is a diagram showing the biodistribution of targeting agents 99mTc:ma-Ser-Ser-Ser-ZHER2342 (99mTc-maSSS-ZHER2342), 99mTc:ma-Glu-Glu- Glu-ZHER2342 (99mTc-maEEE-ZHER2342), 99mTc:ma-Ser-Glu-Glu-ZHER2342 (99mTc- maSEE-ZHER2342), 99mTc:ma-Glu-Glu-Ser-ZHER2342 (99mTc-maEES-ZHER2342), and 99mTc:ma-Glu-Ser-Glu-ZHER2342 (99mTc-maESE-ZHER2342) in Balb/C nu/nu mice bearing SKOV-3 xenografts 4 h p.i.
Figure 9 is a diagram showing the biodistribution of targeting agent 99mTc:ma-Ser-Lys-Ser-ZHER2342 (99mTc-maSKS-ZHER2342) in Balb/C nu/nu mice bearing SKOV-3 xenografts 4 h p.i.
Figure 10 illustrates a gamma-camera image of a mouse bearing HER2-expressing SKOV-3 xenograft using 99mTc:ma-Glu-Ser-Glu-ZHER2342- Arrows point to kidneys (K) and tumors (T). Figure 1 1 illustrates the specificity of uptake of 99mTc:ma-Ser-l_ys-Ser-
ZHER2 342 and 99mTc:ma-Glu-Ser-Glu-ZHER2342 in HER2-expressing tumor xenografts. For blocking, a large molar excess of non-labeled ZHER2 343 was pre-injected in the animals. Data are presented as average percent of injected activity per gram (% IA/g) in four animals ± SEM.
In Figures 1 , 4 and 10, contours of the animals were derived from the digital photographs and superimposed over the gamma-camera images to facilitate interpretation thereof.
In Figures 2-3 and 5-9, data are presented as an average percent of injected activity per gram (% IA/g) of four animals ± SEM. The data for intestines are presented as radioactivity per whole intestines with content, and expressed as % IA per whole sample (typical weight approximately 3 grams).
Example
Outline
In certain forms of breast and ovarian cancer, tumor cells overexpress the protein kinase HER2 on their surfaces. Polypeptides capable of selective interaction with HER2 have been described, such as a monoclonal antibody to HER2 (trastuzumab, marketed as Herceptin® by Genentech and Roche) and engineered combinatorial proteins, for example those designated
ZHER2342 and ZHER2477 (Orlova et al, Cancer Research 66:4339-48 (2006)). In the studies reported below, the HER2 binding polypeptide ZHER2 342 was provided with chelating moieties as described herein, thus forming chelating compounds according to the invention, and with other N3S chelating moieties for comparative purposes.
Peptide synthesis
The HER2 binding molecule ZHER2 342 was synthesized on a 433A Peptide Synthesizer (Applied Biosystems) using standard Fmoc chemistry, essentially as described in Engfeldt et al, Chembiochem 6:1043-50 (2005).
The peptide ZHER2342
(VEN KFN KEM RNAYWEIALLPNLNNQQKRAFI RSLYDDPSQSAN LLAEA-
KKLNDAQAPK; SEQ ID NO:2) was assembled on an Fmoc amide resin with a substitution level of 0.67 mmol/g. The N-terminal Fmoc group was removed by 20 % piperidine-NMP. Acylation reactions were performed in NMP with 10 molar equivalents of amino acid, activated with 2-(1 H-benzothazol-1-yl)- 1 ,1 ,3,3-tetramethyluronium hexafluorophosphate (HBTU) and 1 - hydroxybenzothazole (HOBt) (Advanced ChemTech, Louisville, USA). The 17 underlined residues in the sequence above were double coupled. Unreacted amino groups were capped with acetic anhydride (Applied Biosystems, UK).
The chelating functions ma-Gly-Gly-Gly, ma-Ser-Ser-Ser, ma-Glu-Glu- GIu and ma-Lys-Lys-Lys (all for comparison) and ma-Ser-Glu-Glu, ma-Glu- Ser-Glu, ma-Glu-Glu-Ser and ma-Ser-Lys-Ser (illustrating the invention) were introduced by manual synthesis. The N-terminal Fmoc protecting group was removed by incubation with 20 % piperidine-NMP for 20 min. All manual acylation reactions were performed with 5 molar equivalents of amino acid, HBTU and HOBt and 10 equivalents of N-ethyldiisopropylamine (DIEA, Lancaster Synthesis, UK) in NMP for 20 min. All reactions were monitored by the Kaiser test, and repeated when the yield was found unsatisfactory.
The final deprotection and release of peptides from the resin support was performed as a one-step procedure by treatment with TFA/ethanedithiol(EDT)/thisopropylsilane (TIS)/H2O (94:2.5:2.5:1 ) for 2 h. This was followed by three rounds of extraction in te/t-butyl methyl ether-H2O (50:50) (Merck), filtration and lyophilization. The product was analyzed by analytical RP-HPLC using a 4.6 x 150 mm polystyrene/divenylbenzene matrix column with a particle size of 5 μm (GE Healthcare, Sweden), a 20 min gradient of 20-60 % B (A: 0.1 % TFA-H2O, B: 0.1 % TFA-CH3CN) and a flow rate of 1 ml/min. The peptides were purified using a 20 min gradient of 25-45 % B. The pure product was lyophilized prior to dissolution in 10 mM NH4Ac pH 5,5, and the final peptide concentration was determined by amino acid analysis.
To verify the identity of the peptides, part of the fractions from the HPLC analysis were lyophilized, dissolved in 5 M urea, 0.2 M NH4HCOs, and 30 mM DTT and incubated for 1 h to reduce disulfides before capping free thiol groups with iodoacetamide (0.1 M, 1 h, Sigma, Germany), and finally the peptide mass was analyzed on a Q-Tof™ Il mass spectrometer fitted with an ESI source (Waters Corporation, Micromass MS Technologies, UK). The Maximum Entropy™ 1 (MaxEnti ) algorithm was used to deconvolute isotopic and charge state information.
ZHER2342 was successfully synthesized using Fmoc solid phase peptide synthesis. The synthetic yield was determined to 21 % by analytical RP- HPLC. The total yield of chelating polypeptides was 13-17 %. The peptides were purified to more than 90 % purity. The mass of each peptide was determined by ESI-MS, and the experimentally determined molecular weights correlated well with the theoretically calculated values (Table 2).
Melting point analysis
Variable temperature measurements were performed using a Jasco J- 810 spectropolahmeter (JASCO, Tokyo, Japan). Samples were diluted to a concentration of 50 μM in PBS, pH 7.4. A cell with an optical path length of 1 mm was used. CD spectra from 250 to 195 nm were obtained at 20 0C before and after melting tests. For melting point measurements, the absorbance was measured at 221 nm and the temperature was increased by 5 °C/min from 20 to 90 0C.
All conjugates generated very similar characteristic α-helical spectra. Identical CD spectra were taken before and after a melting point test was performed, indicating that the peptides regained their original fold after melting. The conjugates all had melting points between 64-68 0C, as indicated in Table 2.
Biosensor analysis
Real-time biospecific interaction analysis was used to study the binding kinetics of ZHER2 342 per se and the chelating compounds based on ZHER2342 with chelating moieties on a Biacore 2000 instrument (Biacore, Sweden). Recombinant extracellular domain of HER2 (generous gift from G Adams, Fox Chase Cancer Center, Philadelphia) and the control protein human serum albumin, (HSA, KabiVitrum, Sweden) were diluted to 20 μg/ml in 10 mM NaAc pH 4.5 and immobilized onto a CM5 sensor chip (Biacore). The immobilization level was 500 RU (Response Units) for the HER2 surface and 900 RU for HSA. Samples were diluted in HBS (10 mM HEPES, 150 mM NaCI, 3.4 mM EDTA, 0.005 % Surfactant P20, pH 7.4) to concentrations ranging from 0.3 to 10 nM. The flow rate was set to 50 μl/min with HBS as running buffer, and the samples were injected for 5 min followed by 10 min dissociation and regeneration with 20 μl HCI (20 mM). The BiaEval software (Biacore) and the Langmuir 1 :1 binding model were used for the kinetic analysis.
ZHER2342 per se had an apparent dissociation constant of 76 pM. The corresponding values for the different chelator containing polypeptides proved to be somewhat higher, as shown in Table 2.
Figure imgf000020_0001
Labeling with "171Tc and stability tests
99mTc was obtained as pertechnate by elution of Ultra-TechneKow generator (Tyco, Holland) with sterile 0.9 % NaCI (Mallinckrodt Medical BV, Holland). Before labeling, the freeze-dhed chelating derivatives of ZHER2 342 were dissolved in degassed MiIIi-Q water and stored at -20 0C. Analysis of the labeled derivatives of ZHER2342 was performed using glass fiber sheets impregnated with silica gel for instant thin layer chromatography (ITLC™ SG, Gelman Sciences Inc, USA). To determine labeling yield and stability, ITLS™ SG strips were eluted with PBS. Validation experiments demonstrated that pertechnetate, as well as tartrate, glucoheptonate and cysteine complexes of 99mTc migrate with the eluent front, while derivatives of ZHER2342 do not move under these conditions. To determine the presence of reduced hydrolyzed technetium, ITLC was eluted with pyridine:acetic acid:water (5:3:1.5). When this eluent was used, the technetium colloids stayed, while radiolabeled derivatives of ZHER2342, as well as pertechnetate and tartrate complexes of 99mTc, migrated with the solvent front.
Thawed conjugate solution in ultra-pure water (20 μl, 1 mg/ml) was mixed with 20 μl 0.15 M NaOH, and 10 μl of freshly prepared SnCI2'2H2O solution in 0.01 M HCI (1 mg/ml) was added followed by 200 μl of 99mTc- eluate. The mixture was carefully mixed and incubated at room temperature for 60 min. Use of alkaline conditions gave a labeling yield of approximately 90 % for all conjugates synthesized. The presence of reduced, hydrolyzed technetium was less than 2 % in all experiments.
After labeling, the 99mTc-labeled ZHER2 342 variants were isolated from unreacted technetium and other low-molecular weight components using size-exclusion chromatography with a disposable NAP-5 column (GE Healthcare, Sweden) pre-equilibrated with PBS. Separation was performed according to the manufacturer's instruction. The use of PBS as an eluent made it possible to obtain 99mTc-labeled ZHER2 342 directly in an injectable form after purification. Losses of labeled conjugate during purification were on the order of 13-17 %. According to ITLC analysis of the purified product, its purity was always higher than 95 %. Labels were stable in PBS. Stability tests were performed, in which labeled ZHER2 342 molecules were incubated in PBS or in PBS with 300 molar equivalents of cysteine. Stability was analyzed by ITLC. After a one hour incubation with a 300-fold excess of cysteine at 37 0C, more than 90 % of the radioactivity was associated with the ZHER2 342 molecule, which indicates strong binding of 99mTc. Binding specificity of "mTc-labeled ZHER2342 variants to HER2-expressing SKOV-3 cells
SKOV-3 cells bearing 1.2 x 106 HER2 receptors per cell were cultivated on Petri dishes with a diameter of 3.5 cm to a cell density of 2-5 x 105 CeIIs per dish.
Labeled conjugates, 0.8 ng/105 cells, were added to two groups of Petri dishes containing SKOV-3 cells. One group of dishes was pre-saturated with a 1000-fold excess of non-labeled recombinant ZHER2 342 molecules 10 min before labeled molecules were added. The cells were incubated for 1 h at 37 0C and incubation media was collected. The cell dishes were washed with cold serum-free medium and treated with 0.5 ml trypsin-EDTA solution (0.05 % trypsin, 0.02 % EDTA in PBS buffer, Flow Irvine, UK) for 10 min at 37 0C. When cells were detached, 0.5 ml complete medium was added to every dish and cells were re-suspended. The cell suspension was collected for radioactivity measurements. Cell-associated radioactivity (C) was measured on an automated gamma-counter with a 3-inch NaI(TI) detector (1480 WIZARD, Wallac OY, Finland) in parallel with 1 ml corresponding incubation medium (M). The fraction of added radioactivity bound to cells was calculated as % of bound radioactivity = C x 100 % / (C + M). The results of this binding specificity experiment showed that binding of all 99mTc-labeled ZHER2 342 chelators could be prevented by receptor saturation (data not shown).
Animal studies The animal study was approved by the local Ethics Committee for
Animal Research at Uppsala University, Sweden. In order to create an in vivo tumor model, female outbred Balb/c nu/nu mice (10-12 weeks old at arrival) were used in the in vivo experiments. SKOV-3 tumors were grafted by subcutaneous injection of approximately 7 x 106 cells in the right hind leg. Xenografts were allowed to develop during eight weeks. Biodistribution in normal NMRI mice
To quantify hepatobiliary excretion, groups of four NMRI mice were injected with approximately 1 μg 99mTc-labeled ZHER2342 (approximately 100 kBq). The biodistribution measurement was performed 4 h after injection (pi). Mice were intraperitoneal^ injected with a lethal dose of ketamine HCI
(Ketalar®, Pfizer) and xylazine HCI (Rompun®, Bayer). The mice were killed with heart puncture with a 1 ml syringe rinsed with diluted heparin (Leo Pharma). Organs and tissue samples of blood, lung, liver, spleen, kidney, stomach, salivary glands, thyroid and muscle were excised and collected in weighed plastic bottles. Intestines were collected together with their content. Organs and tissue samples were weighed and their radioactivity was recorded using an automatic gamma-spectrometer with standard 99mTc protocol. The background was measured for each sample holder in the same way. Tissue uptake values were calculated as percent of injected activity per gram tissue (% IA/g).
Radioactivity in kidneys and intestine with contents were selected as a measure of renal and hepatobiliary excretion. Other organs were selected to evaluate in vivo stability of the technetium label. An elevated level of blood radioactivity would be an indication of transchelation of 99mTc to blood plasma proteins, while formation of radio colloids or macroaggregates should be reflected in an elevated uptake in liver, spleen or lungs. Re-oxidation and release of pertechnetate during catabolism of the label should cause increased uptake of radioactivity in stomach, salivary gland and thyroid.
Biodistribution in mice with SKOV-3 xenografts
To study biodistribution in Balb/c nu/nu mice bearing SKOV-3 xenografts, four animals per conjugate were used. For 99mTc:ma-Glu-Ser-Glu- ZHER2342 and 99mTc:ma- Ser-Lys-Ser-ZHER2342, an extra group of four mice was subcutaneously injected with 750 μg of non-labeled recombinant ZHER2 342 in 250 μl PBS 1 h before the radioactive injection (blocking experiment). Mice were subcutaneously injected with the various 99mTc:ma- Xaa1 -Xaa2-Xaa3-ZHER2342 variants in 100 μl PBS (approximately 100 kBq). At 4 h pi, mice were injected intraperitoneal^ with a lethal dose of ketamine HCI (Ketalar®, Pfizer) and xylazine HCI (Rompun®, Bayer). The mice were killed with heart puncture with a 1 ml syringe rinsed with diluted heparin (Leo Pharma). The mice in the blocking group were killed 4 h pi. The samples were treated as in the previously described experiments on biodistribution in normal NMRI mice (see above).
Statistical analysis
An unpaired f-test of the biodistribution data was performed using the GraphPad Prism software (Graph-Pad Software Inc, San Diego, USA). The differences were considered significant if the p values were less than 0.05.
Gamma-camera imaging
Experimental tumor imaging was performed five hours after injection in mice bearing SKOV-3 xenografts. All animals were intravenously injected (tail) with 3 MBq (3 μg) 99mTc:ma-Gly-Gly-Gly-ZHER2342 (comparison), 99mTc:ma-Ser-Ser-Ser-ZHER2342 (comparison) or 99mTc:ma-Glu-Ser-Glu- ZHER2 342 (illustrating the invention). Five hours after injection of the "Relabelled conjugates, all animals were killed by overdosing of ketamine/xylazine. Imaging was performed using a Siemens e.cam gamma- camera equipped with an LEHR collimator. Static images were collected during 10 minutes.
Results pertaining to comparative chelators
Gamma-camera images of mice with HER2-expressing xenografts using 99mTc:ma-Gly-Gly-Gly-ZHER2342 demonstrated capability of this substance to image tumors (Figure 1 ). However, a majority of the radioactivity accumulation was found in the abdomen area. Dissection of imaged animals and subsequent radioactivity measurements showed that this radioactivity was contained in the caecum content. This was considered to be evidence of hepatobiliary excretion of the conjugate. As discussed in the above description of the invention, the hepatobiliary excretion pathway is undesirable, since it generates a high and long-lasting background in the abdomen. The biodistribution experiment in normal mice demonstrated that the use of the ma-Ser-Ser-Ser chelator enabled a reduction of the level of hepatobiliary excretion three-fold, at a cost of moderate increase of radioactivity uptake in the kidneys (Figure 2). The biodistribution experiment in tumor-bearing mice confirmed the capacity of 99mTc:ma-Ser-Ser-Ser-ZHER2342 to target HER2-expressing tumor xenografts. Radioactivity accumulation in the tumors exceeded the accumulation in all organs and tissues, except the kidneys (Figure 3). The tumor accumulation was even higher than the average accumulation in the gastrointestinal tract, with account taken of the fact that the typical sample weight of intestines was 3 gram.
A gamma camera image (Figure 4) demonstrated improvement in contrast in comparison with 99mTc:ma-Gly-Gly-Gly-ZHER2342- Still, a radioactivity accumulation in caecum was clearly seen, which caused a modest contrast between tumor and abdominal area.
When all three amino acids in a chelating ma-Xaa1 -Xaa2-Xaa3 moiety of the ZHER2342 derivatives were lysine or glutamic acid, hepatobiliary excretion was to a high extent suppressed in normal mice (Figure 5). At the same time, a shift to a renal excretion pathway caused an appreciable increase in renal uptake. Kidney accumulation was 95 ± 23 % IA/g (percent injected activity per gram) for 99mTc:ma-Glu-Glu-Glu-ZHER2342 and 127 ± 9 % IA/g for 99mTc:ma-Lys-Lys-Lys-ZHER2342 (compared with 18 ± 4 % IA/g for 99mTc:ma-Ser-Ser-Ser-ZHER2342) Besides, use of ma-Lys-Lys-Lys as a chelator caused high radioactivity uptake in liver, 6.8 ± 2.5 % IA/g (compared with 0.48 ± 0.28 % IA/g for 99mTc:ma-Ser-Ser-Ser-ZHER2342 and 0.21 ± 0.01 % IA/g for 99mTc:ma-Glu-Glu-Glu-ZHER2342).
Results pertaining to chelators of the invention
Unexpectedly, the shift to renal excretion was accompanied by much smaller increase of radioactivity accumulation in kidneys, when only part of serines was substituted by glutamic acids or lysines (Figure 6). Thus, the radioactivity concentration in kidneys for ma-Glu-Glu-Ser and ma-Ser-Glu-Glu was less than half of that of ma-Glu-Glu-Glu, and in the case of ma-Glu-Ser- GIu, the difference was more than four-fold. Apparently, the reduction in renal uptake was not caused by an impaired clearance, since the radioactivity concentration in all other organs and in the rest of the body was at approximately the same level as in the case of ma-Glu-Glu-Glu. Most likely, but without wishing to be bound by this theory, the conjugates were excreted by glomerular filtration, while their re-absorption from primary urine was diminished in comparison with ma-Glu-Glu-Glu.
A similar effect was observed for the ma-Ser-Lys-Ser chelator (Figure 7). This chelator moiety provided as low radioactivity concentration in intestine as ma-Lys-Lys-Lys, but the renal uptake was reduced nearly fourfold (from 127 ± 9 %IA/g to 33 ± 2 %IA/g). Additionally, the use of ma-Ser- Lys-Ser reduced liver uptake twelve-fold in comparison with ma-Lys-Lys-Lys.
The tumor-targeting properties of 99mTc:ma-Glu-Glu-Glu-ZHER2342, 99mTc:ma-Ser-Glu-Glu-ZHER2342, 99mTc:ma-Glu-Glu-Ser-ZHER2342, 99mTc:ma- Glu-Ser-Glu -ZHER2342, and 99mTc:ma-Ser-Lys-Ser-ZHER2342 were verified in mice bearing HER2-expressing SKOV-3 xenografts (Figures 8 and 9). In general, the biodistribution of the inventive chelating compounds was characterized by quick clearance of radioactivity from blood and all organs and tissues. The radioactivity concentration in tumors was higher than in other organs or tissues except for the kidneys. The combination of a reasonably good retention of the radioactive metal atom in tumors on the one hand, and quick blood and whole-body clearance on the other, generated a good contrast (i.e. tumor-to-non-tumor ratio) with respect to a majority of the organs and tissues. As a confirmation of the biodistribution studies, gamma-camera imaging of 99mTc:ma-Glu-Ser-Glu-ZHER2342 (Figure 10) showed that high contrast images of HER2-expressing xenografts in mice can be produced using a conjugate according to the invention. The only organ with apparent accumulation of radioactivity apart from the tumor was the kidney. Though it might create certain problems with detection of HER2-expression in metastases in close proximity to kidneys, the position of kidneys in the body is well-defined, which would help in the interpretation of the gamma-camera images. The reduction in kidney uptake further reduces this problem. Importantly, radioactivity in the abdominal area was very low, and a high contrast of tumor imaging in this area was possible.
To control specificity of tumor uptake, additional groups of animals were pre-injected with a large excess of non-labeled, recombinant ZHER2 342 in order to saturate HER2 receptors in tumor xenografts. Then, 99mTc:ma-Ser- Lys-Ser-ZHER2342 and 99mTc:ma-Glu-Ser-Glu-ZHER2342 were injected. As shown in Figure 11 , injection of a blocking amount of "cold" ZHER2342 reduced significantly the uptake of radioactivity in tumors, thus demonstrating the specificity of the ZHER2342 targeting agent.

Claims

1. Chelating compound of the formula:
ma-Xaa1 -Xaa2-Xaa3-R wherein
- ma is mercaptoacetyl;
- Xaa1 , Xaa2 and Xaa3 are amino acid residues defined as follows: o any one or two of Xaa1 , Xaa2 and Xaa3 is/are the amino acid residue Ser; o in case two of Xaa1 , Xaa2 and Xaa3 are Ser, the remaining amino acid residue is selected from the group consisting of GIu, Arg and Lys; o in case one of Xaa1 , Xaa2 and Xaa3 is Ser, the remaining amino acid residues are both GIu; and
- R has a biological function and is selected from the group consisting of polypeptides, PNA, DNA, RNA, and mixtures thereof.
2. Compound according to claim 1 , in which one or two of Xaa1 , Xaa2 and Xaa3 is/are Ser, and the remaining two or one of Xaa1 , Xaa2 and Xaa3 is/are GIu.
3. Compound according to claim 2, wherein o Xaa3 is GIu; and o the dipeptide Xaa1 -Xaa2 is selected from Ser-Glu, Glu-Ser and Ser-Ser.
4. Compound according to claim 3, wherein o Xaa3 is GIu; and o the dipeptide Xaa1 -Xaa2 is selected from Ser-Glu and Glu-
Ser.
5. Compound according to claim 4, wherein o Xaa1 is Ser; o Xaa2 is GIu; and o Xaa3 is GIu.
6. Compound according to claim 4, wherein o Xaa1 is GIu; o Xaa2 is Ser; and o Xaa3 is GIu.
7. Compound according to claim 3, wherein o Xaa1 is Ser; o Xaa2 is Ser; and o Xaa3 is GIu.
8. Compound according to claim 2, wherein o Xaa3 is Ser; and o the dipeptide Xaa1 -Xaa2 is selected from Ser-Glu, Glu-Ser and GIu-GIu.
9. Compound according to claim 8, wherein o Xaa1 is Ser; o Xaa2 is GIu; and o Xaa3 is Ser.
10. Compound according to claim 8, wherein o Xaa1 is GIu; o Xaa2 is GIu; and o Xaa3 is Ser.
11. Compound according to claim 8, wherein o Xaa1 is GIu; o Xaa2 is Ser; and o Xaa3 is Ser.
12. Compound according to claim 1 , in which two of Xaa1 , Xaa2 and Xaa3 are Ser, and the remaining one of Xaa1 , Xaa2 and Xaa3 is selected from Arg and Lys.
13. Compound according to claim 12, in which two of Xaa1 , Xaa2 and Xaa3 are Ser, and the remaining one of Xaa1 , Xaa2 and Xaa3 is Arg.
14. Compound according to claim 13, wherein o Xaa1 and Xaa2 are Ser; o Xaa3 is Arg.
15. Compound according to claim 13, wherein o Xaa2 and Xaa3 are Ser; o Xaa1 is Arg.
16. Compound according to claim 13, wherein o Xaa1 and Xaa3 are Ser; o Xaa2 is Arg.
17. Compound according to claim 12, in which two of Xaa1 , Xaa2 and Xaa3 are Ser, and the remaining one of Xaa1 , Xaa2 and Xaa3 is Lys.
18. Compound according to claim 17, wherein o Xaa1 and Xaa2 are Ser; o Xaa3 is Lys.
19. Compound according to claim 17, wherein o Xaa2 and Xaa3 are Ser; o Xaa1 is Lys.
20. Compound according to claim 17, wherein o Xaa1 and Xaa3 are Ser; o Xaa2 is Lys.
21. Compound according to claim 12, wherein o Xaa1 and Xaa3 are Ser; o Xaa2 is selected from the group consisting of Arg and Lys.
22. Compound according to any one of claims 1 -21 , wherein said biological function is selected from the group consisting of selective interaction with a target substance; enzymatic action; therapeutic action; and combinations thereof.
23. Compound according to claim 22, wherein said biological function comprises selective interaction with a target substance.
24. Compound according to claim 23, wherein said target substance is selected from the group consisting of molecules bound to a cell surface; molecules bound to intracellular structures; intracellular molecules; components of extracellular matrix; or extracellular molecules.
25. Compound according to claim 24, wherein said target substance is a molecule bound to a cell surface selected from the group consisting of cell surface antigens, cell surface receptors, cell adhesion molecules, and cell surface bound products of a reporter gene.
26. Compound according to claim 25, wherein said molecule bound to a cell surface is selected from the group consisting of PSMA, EpCAM, CEA, GRPR, NMBR, BRS3, CCK1 R, GASR, NTR-1 , NPY-Y2R, GLP1 R, NK1 R, VPAC1 R, VPAC2R, PACR, EDNR, SSR1 -5, TRLR1 -13, uPAR, PPARs, GLUTR, CD antigens including CD4, CD11 a, CD25, CD44, CD52, CD54, CD56, CD64, CD133, CD135, CD213a, CD213b; tyrosine kinase receptors including EGFR, HER2, ERBB3, ERBB4, cMET, RON, Tie1 , Tie2, IGF1 R, IR, antigens specific for neovasculature including VEGFR1 -3, PDGF receptors α and β; G-protein coupled receptors (GPCR) including chemokine receptors; apoptosis related receptors such as TRAILR 1-4, FASR, receptors within the TNFR family; cell adhesion molecules including integrins, cadherins, selectins, desmosomal cadherins, membrane proteoglycans, desmin, syndecan and ion channels.
27. Compound according to claim 24, wherein said target substance is a molecule bound to an intracellular structure, which molecule is selected from the group consisting of cytoskeletal proteins, keratin family, lamins, vimentin, desmin, actin, tubulin, GFAP and melanin.
28. Compound according to claim 24, wherein said target substance is an intracellular molecule selected from the group consisting of kinases, phosphatases and transcription factors, including members of the MAPK kinase, JAK-STAT, EGFR, VEGFR, insulin, integrin, mTOR, NF-κB, Notch, P53, Wnt, TGFb, TNFRs, ToIIR, FAK and phosphatidylinositol signalling pathways, molecules involved in apoptosis including members of the caspase cascade and those involved in the mitochondrial pathway for apoptosis and cell cycle progression including cyclins and cyclin dependent kinases, as well as protein regulatory molecules including the SOCS box containing proteins, ubiquitinating enzymes, sumoylating proteins, the proteasome complex, heat shock proteins and melanin.
29. Compound according to claim 24, wherein said target substance is an extracellular molecule selected from the group consisting of cytokines, such as members of the TNF and IFN families, interleukins and colony stimulating factors, chemokines including the C, CC, CXC and CX3C families, hormones, such as oestrogen, GH, PRL, erythropoietin, activin, TSH, LSH, ACTH, melatonin, NPY, gastrin, and growth factors, such as FGFs, PDGFs, VEGFs, IGFI s, TGFb, EGF, Heregulin and Neuregulins, plasma proteins, including enzymatically active proteins or inhibitors such as the MMP, ADAM and serpin families, high abundant serum proteins, complement factors, hormone and cytokine binding factors and proteins involved in lipid transportation.
30. Compound according to claim 24, wherein said target substance is a component of extracellular matrix selected from the group consisting of collagen and laminin families, fibronectin, elastin, endostatins, thrombospondins, tenascin and fibulin.
31. Compound according to any preceding claim, wherein R is a polypeptide.
32. Compound according to claim 31 , wherein said polypeptide has from 3 to 200 amino acid residues.
33. Compound according to claim 32, wherein said polypeptide has from 6 to 120 amino acid residues.
34. Compound according to claim 33, wherein said polypeptide has from 30 to 70 amino acid residues.
35. Compound according to claim 32, wherein said polypeptide has from 3 to 30 amino acid residues.
36. Compound according to claim 35, wherein said polypeptide has from 3 to 10 amino acid residues.
37. Compound according to claim 36, wherein said polypeptide has 8 amino acid residues.
38. Compound according to any one of claims 35-37, R comprises a polypeptide which is selected from peptide targeting agents based on somatostatin, bombesin, CCK, neurotensin, NPY, GLP1 and NK1.
39. Compound according to any one of claims 23-38, wherein R comprises a binding polypeptide selected from the group consisting of an antibody, an antibody fragment and an antibody derivative.
40. Compound according to any one of claims 23-38, wherein R comprises a binding polypeptide derived from a non-antibody scaffold.
41. Compound according to claim 40, wherein said scaffold is selected from the group consisting of three-helix domains, lipocalins, ankyrin repeat domains, cellulose binding domains, Y crystallins, green fluorescent protein, human cytotoxic T lymphocyte-associated antigen 4, protease inhibitors, PDZ domains, peptide aptamers, staphylococcal nuclease, tendamistats, fibronectin type III domain, zinc fingers, avimers, microproteins and conotoxins.
42. Compound according to claim 41 , wherein said scaffold is a three- helix polypeptide domain.
43. Compound according to claim 42, wherein said scaffold is based on any three-helix domain of Protein G from Streptococcus spp.
44. Compound according to claim 43, wherein said scaffold is the albumin binding domain of Protein G from Streptococcus pyogenes strain G418.
45. Compound according to claim 42, wherein said scaffold is based on any domain of Protein A from Staphylococcus aureus.
46. Compound according to claim 45, wherein said scaffold is the protein Z variant of the B domain of Protein A from Staphylococcus aureus.
47. Compound according to any one of claims 1 -32, wherein R comprises a polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NO:1-12 in Table 1.
48. Compound according to any one of claims 1 -32, wherein R comprises a polynucleotide molecule selected from DNA, RNA, PNA and mixtures thereof.
49. Compound according to claim 48, wherein said polynucleotide molecule comprises a DNA molecule.
50. Compound according to claim 48, wherein said polynucleotide molecule comprises an RNA molecule.
51. Compound according to claim 48, wherein said polynucleotide molecule comprises a PNA molecule.
52. Compound according to any one of claims 48-51 , wherein said polynucleotide molecule comprises an aptamer.
53. Compound according to any one of claims 48-51 , wherein said polynucleotide molecule comprises an antisense polynucleotide.
54. Chelate of a compound according to any one of claims 1 -53 with a radioactive moiety selected from M, M=O and O=M=O, wherein M is a radioactive metal atom.
55. Chelate according to claim 54, wherein M is suitable for in vivo imaging applications.
56. Chelate according to claim 55, wherein M is selected from the group consisting of 99mTc, 94Tc and 96Tc.
57. Chelate according to claim 56, wherein M is 99mTc.
58. Method of preparing a chelate according to any one of claims 54- 57, comprising mixing, in the presence of a reducing agent, of a compound according to any one of claims 1 -53 with a radioactive moiety MO4 ", wherein M is a radioactive metal atom.
59. Method of preparing a chelate according to any one of claims 55- 56, comprising mixing, in the presence of a reducing agent, of a compound according to any one of claims 1 -53 with a compound [""1Tc]TcO4 ", [94Tc]TcO4 " and [96Tc]TcO4 ".
60. Method of preparing a chelate according to claim 57, comprising mixing, in the presence of a reducing agent, of a compound according to any one of claims 1 -53 with [99mTc]TcO4\
61. Method according to any one of claims 58-60, in which said reducing agent is selected from the group consisting of SnCb and SnF2.
62. Method according to any one of claims 58-61 , in which said mixing is carried out in the presence of a weak chelator, which acts as an intermediate in transfer of M to the compound according to any one of claims 1 -53.
63. Method according to claim 62, in which said weak chelator is selected from the group consisting of tartrate, glucoheptonate and citrate.
64. Method of in vivo imaging of the body of a mammalian, including human, subject, comprising the steps: - administering a chelate according to any one of claims 55-57 into the body of a mammalian subject; and - obtaining an image of at least a part of the subject's body using a medical imaging instrument, said image indicating the presence of M inside said body.
65. Method according to claim 64, in which the step of obtaining an image is repeated at least three times, whereby a series of images is obtained.
66. Method according to any one of claims 64-65, comprising, before the administration step, a step of preparing a chelate according to any one of claims 55-56, comprising mixing, in the presence of a reducing agent, of a compound according to any one of claims 1 -53 with a compound selected from [99mTc]TcO4 ", [94Tc]TcO4 " and [96Tc]TcO4 ".
67. Method according to any one of claims 64-65, comprising, before the administration step, a step of preparing a chelate according to claim 57, comprising mixing, in the presence of a reducing agent, of a compound according to any one of claims 1 -53 with [""1Tc]TcO4 ".
68. Chelate according to any one of claims 55-57 for use in diagnostics.
69. Use of a chelate according to any one of claims 55-57 in the preparation of a diagnostic agent for imaging in vivo of the body of a mammalian, including human, subject.
PCT/EP2007/061438 2006-10-27 2007-10-24 New chelating compound WO2008049866A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP06123095 2006-10-27
EP06123095.9 2006-10-27
US85550106P 2006-10-31 2006-10-31
US60/855,501 2006-10-31

Publications (1)

Publication Number Publication Date
WO2008049866A1 true WO2008049866A1 (en) 2008-05-02

Family

ID=38917642

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/061438 WO2008049866A1 (en) 2006-10-27 2007-10-24 New chelating compound

Country Status (1)

Country Link
WO (1) WO2008049866A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012087908A1 (en) * 2010-12-22 2012-06-28 Ge Healthcare Limited Her2 binding peptides labeled with aluminium-[18] fluoride complexed by nota
WO2012087912A1 (en) * 2010-12-22 2012-06-28 Ge Healthcare Limited Her2 binding peptides labelled with a 18f - containing organosilicon compound
CN102827208A (en) * 2012-09-25 2012-12-19 北京师范大学 Preparation method and application of 99mTcO-core-labeled methionine dithiocarbamate complex
US9061080B2 (en) 2010-12-22 2015-06-23 Ge Healthcare Limited HER2 binding peptides labeled with aluminium-[18] fluoride complexed by NOTA
RU2655965C2 (en) * 2016-10-12 2018-05-30 Федеральное государственное бюджетное образовательное учреждение высшего образования "Сибирский государственный медицинский университет" Министерства здравоохранения Российской Федерации (ФГБОУ ВО СибГМУ Минздрава России) Method of obtaining set of technetium-99m complex with the modified specific mini-antibodies for diagnostics of oncological diseases with her2/neu overexpression
US10894097B2 (en) 2008-12-22 2021-01-19 General Electric Company HER2 binders
CN115028736A (en) * 2022-05-10 2022-09-09 南京大学 Targeting molecular probe and application

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DECRISTOFORO C ET AL: "THE INFLUENCE OF CHELATOR ON THE PHARMACOKINETICS OF 99MTC-LABELLED PEPTIDES", QUARTERLY JOURNAL OF NUCLEAR MEDICINE, MILAN, IT, vol. 46, no. 3, 2002, pages 195 - 205, XP008051358, ISSN: 1125-0135 *
ZHU Z ET AL: "A novel and simplified route to the synthesis of N3S chelators for <99m>Tc labeling<1>", NUCLEAR MEDICINE AND BIOLOGY, ELSEVIER, NY, US, vol. 28, no. 6, August 2001 (2001-08-01), pages 703 - 708, XP004300998, ISSN: 0969-8051 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10894097B2 (en) 2008-12-22 2021-01-19 General Electric Company HER2 binders
US20130295010A1 (en) * 2010-12-22 2013-11-07 Duncan Hiscock Her2 binding peptides labelled with a 18f - containing organosilicon compound
WO2012087912A1 (en) * 2010-12-22 2012-06-28 Ge Healthcare Limited Her2 binding peptides labelled with a 18f - containing organosilicon compound
US11633507B2 (en) 2010-12-22 2023-04-25 General Electric Company HER2 binders
WO2012087908A1 (en) * 2010-12-22 2012-06-28 Ge Healthcare Limited Her2 binding peptides labeled with aluminium-[18] fluoride complexed by nota
RU2592685C2 (en) * 2010-12-22 2016-07-27 Дженерал Электрик Компани Radioactive labelled peptides binding to her2
JP2014503546A (en) * 2010-12-22 2014-02-13 ゼネラル・エレクトリック・カンパニイ Radiolabeled HER2 binding peptide
WO2012096760A1 (en) * 2010-12-22 2012-07-19 General Electric Company Radiolabled her2 binding peptides
US9061080B2 (en) 2010-12-22 2015-06-23 Ge Healthcare Limited HER2 binding peptides labeled with aluminium-[18] fluoride complexed by NOTA
CN103491984A (en) * 2010-12-22 2014-01-01 通用电气公司 Radiolabled her2 binding peptides
AU2011354599B2 (en) * 2010-12-22 2016-09-29 Affibody Ab Radiolabled HER2 binding peptides
EP3936155A1 (en) * 2010-12-22 2022-01-12 General Electric Company Radiolabeled her2-binding peptide conjugates
CN102827208B (en) * 2012-09-25 2015-01-14 北京师范大学 Preparation method and application of 99mTcO-core-labeled methionine dithiocarbamate complex
CN102827208A (en) * 2012-09-25 2012-12-19 北京师范大学 Preparation method and application of 99mTcO-core-labeled methionine dithiocarbamate complex
RU2655965C2 (en) * 2016-10-12 2018-05-30 Федеральное государственное бюджетное образовательное учреждение высшего образования "Сибирский государственный медицинский университет" Министерства здравоохранения Российской Федерации (ФГБОУ ВО СибГМУ Минздрава России) Method of obtaining set of technetium-99m complex with the modified specific mini-antibodies for diagnostics of oncological diseases with her2/neu overexpression
CN115028736A (en) * 2022-05-10 2022-09-09 南京大学 Targeting molecular probe and application
CN115028736B (en) * 2022-05-10 2023-07-25 南京大学 Targeting molecular probe and application thereof

Similar Documents

Publication Publication Date Title
JP6959616B2 (en) Site-specific RI-labeled antibody with IgG-binding peptide
Tran et al. 99mTc-maEEE-ZHER2: 342, an Affibody molecule-based tracer for the detection of HER2 expression in malignant tumors
Malmberg et al. Comparative evaluation of synthetic anti-HER2 Affibody molecules site-specifically labelled with 111 In using N-terminal DOTA, NOTA and NODAGA chelators in mice bearing prostate cancer xenografts
WO2008049866A1 (en) New chelating compound
Tran et al. In vivo evaluation of cysteine-based chelators for attachment of 99mTc to tumor-targeting Affibody molecules
JP7327762B2 (en) Modified antibodies and radiometal-labeled antibodies
TWI795415B (en) A novel anti-human CEACAM5 antibody Fab fragment
Honarvar et al. Evaluation of the first 44Sc-labeled Affibody molecule for imaging of HER2-expressing tumors
JP7036996B2 (en) RI-labeled humanized antibody
Tran et al. Effects of lysine-containing mercaptoacetyl-based chelators on the biodistribution of 99mTc-labeled anti-HER2 Affibody molecules
von Witting et al. Selection of the optimal macrocyclic chelators for labeling with 111In and 68Ga improves contrast of HER2 imaging using engineered scaffold protein ADAPT6
Altai et al. Preclinical evaluation of anti-HER2 Affibody molecules site-specifically labeled with 111In using a maleimido derivative of NODAGA
CN110339375B (en) HER 2-targeted rk polypeptide radiopharmaceuticals and preparation method thereof
Morais et al. Evaluation of novel 99m Tc (I)-labeled homobivalent α-melanocyte-stimulating hormone analogs for melanocortin-1 receptor targeting
Garousi et al. Radionuclide therapy using ABD-fused ADAPT scaffold protein: Proof of Principle
Orlova et al. Pre-clinical evaluation of [111In]-benzyl-DOTA-ZHER2: 342, a potential agent for imaging of HER2 expression in malignant tumors
Rudd et al. Enzyme mediated incorporation of zirconium-89 or copper-64 into a fragment antibody for same day imaging of epidermal growth factor receptor
Farzin et al. Clinical aspects of radiolabeled aptamers in diagnostic nuclear medicine: A new class of targeted radiopharmaceuticals
Ui et al. Development and characterization of a 68Ga-labeled A20FMDV2 peptide probe for the PET imaging of αvβ6 integrin-positive pancreatic ductal adenocarcinoma
Rosik et al. Direct comparison of 111 In-labelled two-helix and three-helix Affibody molecules for in vivo molecular imaging
Okarvi et al. Preparation and evaluation of the tumor-specific antigen-derived synthetic mucin 1 peptide: A potential candidate for the targeting of breast carcinoma
Ballard et al. In vitro and in vivo evaluation of melanin-binding decapeptide 4B4 radiolabeled with 177Lu, 166Ho, and 153Sm radiolanthanides for the purpose of targeted radionuclide therapy of melanoma
ES2840323T3 (en) Tumor Targeting Peptide Variants
Tolmachev et al. Influence of an aliphatic linker between DOTA and synthetic ZHER2: 342 Affibody molecule on targeting properties of the 111In-labeled conjugate
TW200812627A (en) Compounds as aptamer-dimers and their uses in diagnosis and therapy

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: 07821801

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07821801

Country of ref document: EP

Kind code of ref document: A1