WO2010066051A1 - Thiol-nota derivatives for kit 68ga radiolabeling - Google Patents

Thiol-nota derivatives for kit 68ga radiolabeling Download PDF

Info

Publication number
WO2010066051A1
WO2010066051A1 PCT/CA2009/001822 CA2009001822W WO2010066051A1 WO 2010066051 A1 WO2010066051 A1 WO 2010066051A1 CA 2009001822 W CA2009001822 W CA 2009001822W WO 2010066051 A1 WO2010066051 A1 WO 2010066051A1
Authority
WO
WIPO (PCT)
Prior art keywords
nota
thiol
protein
formula
derivative
Prior art date
Application number
PCT/CA2009/001822
Other languages
French (fr)
Inventor
Raif Schirrmacher
Carmen Waengler
Björn WAENGLER
Original Assignee
The Royal Institution For The Advancement Of Learning/Mcgill University
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 The Royal Institution For The Advancement Of Learning/Mcgill University filed Critical The Royal Institution For The Advancement Of Learning/Mcgill University
Publication of WO2010066051A1 publication Critical patent/WO2010066051A1/en

Links

Classifications

    • 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
    • A61K51/0474Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group
    • A61K51/0482Organic compounds complexes or complex-forming compounds, i.e. wherein a radioactive metal (e.g. 111In3+) is complexed or chelated by, e.g. a N2S2, N3S, NS3, N4 chelating group chelates from cyclic ligands, e.g. DOTA
    • 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
    • A61K51/081Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins the protein being an albumin, e.g. human serum albumin [HSA], bovine serum albumin [BSA], ovalbumin
    • 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
    • A61K51/087Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins the peptide being an annexin, e.g. annexin V
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D255/00Heterocyclic compounds containing rings having three nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D249/00 - C07D253/00
    • C07D255/02Heterocyclic compounds containing rings having three nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D249/00 - C07D253/00 not condensed with other rings

Definitions

  • the present disclosure broadly relates to a composition of matter for introducing 68 Ga into proteins and peptides. More specifically, but not exclusively, the present disclosure relates to novel 1 ,4,7-triazacyclononane- 1,4,7- triacetic acid (NOTA) derivatives. The present disclosure also relates to Ga radiolabeled l,4,7-triazacyclononane-l,4,7-triacetic acid (NOTA) derivatives. Moreover, the present disclosure also relates to the use of such radiolabeled derivatives for positron emission tomography (PET) imaging.
  • PET positron emission tomography
  • radionuclides for PET imaging include C, F,
  • Bioactive molecules labeled with radionuclides suitable for molecular imaging (e.g. PET) purposes are of increasing interest.
  • Ga has gained in popularity as it enables the fast, simple and convenient labeling of peptides and other small biologically active materials such as antisense oligonucleotides. Furthermore, the labeling with 68 Ga is independent from cyclotrons and is thus highly flexible as it can be readily generated from a 68 Ge/ 68 Ga generator system.
  • 68 Ga 3+ was mixed with the peptide-chelator-conjugate DOTATOC and kept at pH 4.8 and a temperature of 9O 0 C over a period of 15 min (conditions that would denature any protein).
  • the radiochemical yield of 68 Ga- DOTA-RGD was shown to be approximately 60% as compared to approximately 30% for 18 F-galacto-RGD. While tumor uptake ratios were comparable, tumor/blood ratios were higher for 111 In-DOTA-RGD and 18 F-galacto-RGD than for 68 Ga-DOTA- RGD, largely due to a higher blood pool activity ⁇ i.e. higher background activity).
  • the lack of stability of 68 Ga- DOTA-RGD was shown to be approximately 60% as compared to approximately 30% for 18 F-galacto-RGD. While tumor uptake ratios were comparable, tumor/blood ratios were higher for 111 In-DOTA-RGD and 18 F-galacto-RGD than for 68 Ga-DOTA- RGD, largely due to a higher blood pool activity ⁇ i.e. higher background activity).
  • DOTA-peptides could be overcome by using alternative chelator systems such as p- SCN-Bn-NOTA, which could also be better suited for the clinical use of gallium [3]. While the currently used NOTA derivatives are well-suited for the derivatization of peptides, they do not yet allow the derivatization of proteins.
  • NODAGA( ⁇ BU) 3 The cleavage of the protecting groups of the monoreactive NOTA derived prochelator NODAGA( ⁇ BU) 3 (4-(4,7-bis(2-tert- butoxy-2-oxoethyl)- 1 ,4,7-triazonan- 1 -yl)-5-ter/-butoxy-5-oxopentanoic acid) requires concentrated trifluoroacetic acid [4].
  • NODAGA(tBu) 3 was synthesized in five steps in an overall yield of 21% and was subsequently coupled to the peptide [Tyr3] -octreotide (TOC) providing the peptide-chelator conjugate NODAGATOC.
  • the conjugate was used as a SPECT and PET tracer when labeled with 111 In, 67 Ga, or 68 Ga.
  • NOTA-RGDl NOTA-c(RGDyK)
  • NOTA-RGD2 NOTA-E[c(RGDyK)]2
  • N0TA-RGD4 NOTA-E ⁇ E[c(RGDyK)]2 ⁇ 2
  • Radioactive Ga-NOTA-RGD peptides were purified by semi-preparative HPLC leading to decay-corrected yields of 90% for NOTA-RGDl, 82% for NOTA-RGD2 and 64% for N0TA-RGD4.
  • the specific activity of the 68 Ga-NOTA-RGD multimers was measured to be about 9.7-13.6 MBq/nmol. Similar results have been published [6]. [0009] Velikyan et al. have previously reported a straightforward labeling method for quickly introducing macrocyclic bifunctional chelators into Ga peptide-chelator conjugates at room temperature as demonstrated for NODAGA- TATE in which NOTA has been coupled to the eight amino acid residue peptide [Tyr3]Octreotate [7].
  • the purification of the 68 Ga-labeled products was described as being unnecessary since the radiochemical purity was >95%.
  • NOTA derivatives such as TACN-TM (l,4,7-tris(2-mercaptoethyl)-l,4,7- triazacyclononane), NODASA (1,4,7-tri-azacyclononane-l -succinic acid-4,7- deacetic acid), NOTP (l,4,7-triazacyclononane-N,N',N"- tris(methylenephosphonic)acid), and NOTPME (l,4,7-triazacyclononane-N,N',N"- tris(methyl-enephosphonate-monoethylester)) were shown to demonstrate high thermodynamic stability and similar plasma and in vivo stability.
  • TACN-TM l,4,7-tris(2-mercaptoethyl)-l,4,7- triazacyclononane
  • NODASA 1,4,7-tri-azacyclononane-l -succinic acid-4,7- deacetic acid
  • NOTP l
  • the p-SCN-Bn-NOTA chelator has an additional inherent disadvantage. While only a limited number of chelators can be introduced into peptides, proteins might have more than one site capable of accepting a chelator. While an increased number of chelators per macromolecule could increase the specific activity of the chelator-protein conjugate, it has been shown that a high number of derivatization sites leads to a dramatic loss of the biological activity of the derivatized biomolecule [15, 16, 17]. In case of the isothiocyanate derivative of NOTA, the number of derivatization sites per protein molecule could only be determined by means of 14 C labeled chelators or isotopic dilution titration which represent highly complex methods.
  • the present disclosure broadly relates to thiol-NOTA derivatives.
  • the present disclosure relates to a thiol-
  • n 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • m is 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the present disclosure relates to a thiol-
  • the present disclosure broadly relates to Ga radiolabeled thiol-NOTA derivatives.
  • the present disclosure relates to a 68 Ga radiolabeled thiol-NOTA derivative of Formula II:
  • n 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
  • m is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
  • the present disclosure relates to a 68 G/ a radiolab led thiol-NOTA derivative of Formula Ha:
  • the present disclosure broadly relates to thiol-NOTA derivatives for protein and peptide labeling.
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for protein and/or peptide labeling.
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for protein labeling.
  • the present disclosure relates to the se of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling hMAb 425.
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling human serum albumin (HSA).
  • HSA human serum albumin
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling rat serum albumin (RSA).
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling diphtheria toxin (CRM mutant).
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling human annexin V.
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia.
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and hMAb 425.
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and human serum albumin (HSA).
  • HSA human serum albumin
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and rat serum albumin (RSA).
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and diphtheria toxin (CRM mutant).
  • the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and human annexin V.
  • the present disclosure relates to
  • radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and hMAb 425.
  • the present disclosure relates to
  • radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and human serum albumin (HSA).
  • the present disclosure relates to
  • radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and rat serum albumin (RSA).
  • the present disclosure relates to
  • radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and bovine serum albumin (BSA).
  • the present disclosure relates to
  • Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and diphtheria toxin (CRM mutant).
  • the present disclosure relates to
  • Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and human annexin V.
  • the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula IIa for peptide labeling.
  • the present disclosure relates to the use of 68 Ga-radiolabelled thiol-NOTA derivatives of Formula II and Formula Ha for positron emission tomography (PET) imaging.
  • the present disclosure relates to a
  • the present disclosure relates to a
  • 68 Ga protein labeling kit comprising a thiol-NOTA derivative of Formula I or Formula Ia and a maleimide-based linker for conjugating the thiol-NOTA derivative of Formula I or Formula Ia to a target protein.
  • the present disclosure relates to a
  • 68 Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and a target protein.
  • the present disclosure relates to a
  • Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and hMAb 425.
  • the present disclosure relates to a
  • Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and human serum albumin (HSA).
  • HSA human serum albumin
  • the present disclosure relates to a
  • 68 Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and rat serum albumin (RSA).
  • RSA rat serum albumin
  • 68 Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • the present disclosure relates to a
  • 68 Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and diphtheria toxin (CRM mutant).
  • the present disclosure relates to a
  • 68 Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and human annexin V.
  • the present disclosure relates to a method for 68 Ga radiolabeling of a target protein, the method comprising:
  • the present disclosure relates to a method for Ga radiolabeling of a target protein, the method comprising: [0063] reacting a thiol-NOTA derivative of Formula I or Formula Ia with a maleimide containing reagent to afford a maleimide-modified thiol-NOTA conjugate;
  • FIG. 1 shows HPLC chromatograms illustrating the radioactivity traces for labeling mixtures comprising 68 Ga-labeled thiol-NOTA- annexin (A), 68 Ga-labeled thiol-NOTA-diphtheria toxin CRM mutant (B) and 68 Ga- labeled thiol-NOTA-RSA (C).
  • the 68 Ga-labeled conjugates were produced using thiol-NOTA derivative 3 (Formula Ia) and linker 5.
  • FIG. 2 shows a maximum intensity projection image of a blood pool scan of a healthy rat with Ga-labeled thiol-NOTA-RSA acquired over 30 min, demonstrating radiotracer accumulation in large arteries, the liver, and the heart. Atria and ventricles can be easily identified.
  • RA right atrium
  • LA left atrium
  • RV right ventricle
  • LV left ventricle
  • RK right kidney
  • LK left kidney
  • B bladder.
  • FIG. 3 shows a maximum intensity projection image of an anterior myocardial infarction in a rat with [ 18 F] Fluorodeoxyglucose (FDG) (leftside image) and with 68 Ga-labeled thiol-NOTA-Annexin V acquired over 60 min (right-side image).
  • the middle image is an overlay of the left and right-side images.
  • the right-side image demonstrates the superiority of direct labeling of apoptotic tissue via apoptosis specific Annexin-V versus negative demonstration by the use of FDG which shows no uptake into apoptotic/necrotic tissue.
  • the present disclosure relates to thiol-NOTA derivatives for protein and peptide labeling.
  • the thiol-NOTA derivatives of the present disclosure function as chelators for Ga.
  • the thiol-NOTA derivatives of Formula I or Formula Ia are coupled to a target protein using a maleimide-based linker.
  • a target protein is modified using 4-(N-maleimidomethyl)cyclohexane-l-carboxylic acid 3- sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) to produce a maleimide modified protein.
  • sulfo-SMCC 4-(N-maleimidomethyl)cyclohexane-l-carboxylic acid 3- sulfo-N-hydroxysuccinimide ester sodium salt
  • suitable maleimide-based linkers are known in the art and are within the capacity of a skilled technician.
  • a non-limiting example of such other suitable maleimide-based linkers includes 1 ,2-bis(maleimido)ethane.
  • the maleimide modified proteins exhibited a modified molecular size as well as a different structure when compared to the starting protein.
  • the maleimide modified proteins are subsequently coupled with the thiol-NOTA derivatives of Formula I or Formula Ia.
  • NOTA derivatives of Formula I or Formula Ia are important. This observation is further compounded when working with proteins of limited availability and high cost. In an embodiment of the present disclosure, deprotection steps, prior and following protein coupling, are eliminated. Moreover, the number of derivation sites per protein should be controlled such that coupling does not entail loss of biological activity.
  • the thiol-NOTA derivatives of the present disclosure afford high coupling rates at ambient temperatures with maleimide modified proteins leading to protein conjugates of high chemical purity.
  • the thiol-NOTA-protein conjugates are obtained in yields ranging from 85-99%.
  • the maleimide-modified proteins comprise a well-defined number of derivatization sites for coupling with the thiol-NOTA derivatives of Formula I or Formula Ia.
  • the thiol-NOTA protein conjugates (labeling precursors) are readily labeled with Ga to provide Ga-radiolabelled thiol-NOTA-protein conjugates. The radiolabeling process does not affect the biological activity of the target protein.
  • the thiol-NOTA-protein conjugates are labeled with Ga within a period of time ranging from about 5 to about 10 minutes.
  • the thiol-NOTA- protein conjugates are labeled with 68 Ga in high specific radioactivities ranging from about 10 to about 50 GBq/ ⁇ mol.
  • the thiol-NOTA-protein conjugates are labeled with 68 Ga in radiochemical yields >95%.
  • the 68 Ga-radiolabeled thiol-NOTA-protein conjugates of the present disclosure are stable (i.e. radiochemical yields remain substantially constant).
  • thiol-NOTA protein conjugate in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 2.
  • Target protein (4) is derivatized using 4-(N-maleimidomethyl)cyclo-hexane-l- carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) (5) to provide maleimide-modified protein (6).
  • thiol- NOTA derivative 3 (Formula Ia) provides the thiol-NOTA-protein conjugate labeling precursor (7).
  • the target protein was modified using 4-(N- maleimidomethyl)cyclohexane- 1 -carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) to produce a maleimide modified protein.
  • the maleimide group provides for covalent thiol-maleimide coupling at ambient temperatures within a period of time ranging from about 5 to about 10 minutes.
  • the number of maleimide groups introduced into the target protein for subsequent thiol- NOTA coupling could be quantified using Ellman's assay.
  • Target proteins e.g.
  • maleimide modified proteins comprising from about 1.4 to about 1.7 maleimide groups/protein. This results in only minor alterations of the biological activity of the target protein [15, 16, 17].
  • the maleimide-modified proteins were purified using size-exclusion gel chromatography (recovery rates of 95-98%).
  • the modification of recombinant target proteins with free cysteine residues provides an additional improvement.
  • the newly introduced cysteine residue(s) allow(s) for the preparation of a maleimide modified protein with a site specific instead of a limited but random modification site.
  • the thiol-NOTA-protein conjugates were radiolabeled with 68 Ga by mixing each protein conjugate (6.9 nmol) with 1 mL of Ga (240-340 MBq, eluted from a 6 Ge/ 68 Ga generator) in an acetate-buffered aqueous medium at ambient temperature (pH ranging from 3.5 to 4.0) over a period ranging from 5 to 10 minutes (Scheme 3). All labeling reactions were complete after a period of time ranging from 7-10 minutes affording the corresponding Ga radiolabeled thiol- NOTA-protein-conjugates in suitable radiochemical purities.
  • the thiol-NOTA-protein-conjugates were shown to be stable in aqueous solution for at least 4 weeks at 4°C as they could be labeled with unvarying radiochemical yields.
  • the thiol-NOTA derivative of Formula Ia was demonstrated to be stable for 2 years at room temperature.
  • Size-exclusion gel chromatography was carried out using NAP-5 or NAP-10 columns (GE Healthcare Bio-Sciences AB, Uppsala, Sweden) and size-exclusion fast protein liquid chromatography (FPLC) was performed using a Superdex 200 10/30 GL column (Amersham Biosciences AB, Uppsala, Sweden). Ultrafiltration was performed using Vivaspin 6 centrifugal concentrators (Sartorius, G ⁇ ttingen, Germany) with a MWCO of 10 or 100 kDa.
  • a freshly prepared solution of thiol-NOTA (3, 131 ⁇ g; 300 nmol) was added to a solution comprising a maleimide-modified protein in phosphate buffer (0.1M, pH 7.2) and stirred over a period of 30 minutes.
  • the proteinaceous solution was subsequently diluted with HEPES buffer (0.025 M, pH 4.0) and purified by ultrafiltration applying Vivaspin 6 centrifugal concentrators (MWCO 10 IcDa for annexin V, HSA, BSA, RSA and diphtheria toxin (CRM mutant); MWCO 100 kDa for hMAb 425). The ultrafiltration was repeated four times.
  • the thiol-NOTA-protein conjugates were recovered at high yields (85-99%).
  • HEPES buffer (0.025M, pH 4.0) was added to 240-340 MBq of 68 Ga in a sodium acetate solution (1 mL; 0.11 M; pH 3.5-4.0) and incubated for 5 to 10 minutes. The reaction mixtures were subsequently analyzed by analytical HPLC. The Ga- radiolabeled thiol-NOTA-protein conjugates were found to be 95-99% pure and obtained in specific radioactivities ranging from 10 and 50 GBq/ ⁇ mol.
  • CD rats were housed according to German animal protection laws and protocols of the local committee. Experiments were performed under inhalation anesthesia (oxygen 1.2 L/min, isofluran 1.5 vol %) and appropriate warming to prevent hypothermia.
  • Ga-radiolabeled thiol-NOTA-RSA provides a blood pool image of a healthy rat including a clear illustration of the heart, aorta, arteries, and the liver.
  • radioactivity accumulation can be seen in the kidneys and the bladder due to metabolization and excretion processes.
  • the left coronary artery was ligated to induce transmural myocardial infarction. Under anesthesia with intramuscular administration of midazolam (0.1 mg/kg), fentanyl (1 mg/kg), and medetomidin (10 mg/kg) (MMF) and mechanical ventilation, the chest was opened to expose the heart. A 7-0 polypropylene suture on a small curved needle was passed through the LCA and ligated to occlude the LCA. To assess the relationship of in vivo and ex vivo measurements in acute and subacute phases, animals were allowed 2 days of recovery before PET imaging.
  • Annexin V was used as an indicator for apoptotic tissue, as can be found in infarcted tissue. While the imaging of a healthy heart with [ 18 F] Fluorodeoxyglucose (FDG) would demonstrate a V-shaped structure, this shape is interrupted in an infarcted heart due to apoptotic tissue that can no longer absorb any nutrients, (i.e. glucose). Contrary to the lack of FDG in the apoptotic infarcted tissue, the use of annexin V as a marker for apoptosis can directly depict the infarcted site.
  • FDG Fluorodeoxyglucose
  • FIG. 3 shows the overlay of two maximum intensity projection images of an anterior myocardial infarction in a rat.
  • the image covers a cross section through the coronary level of the heart.
  • Negative landmarking of the infarct region was obtained by the use of [ 18 F] Fluorodeoxyglucose (FDG) while 68 Ga- radiolabelded annexin V allows positive infarct imaging.
  • FDG landmarking indicates the infarct region by the lack of accumulating FDG depicted in colors ranging from yellow to red, i.e. the bright apical dotted structure and the interior light and medium grey horizontal C-shaped structure.
  • Ga-radiolabeled annexin V The direct accumulation of the Ga-radiolabeled annexin V in the apoptotic and necrotic infarct heart tissue is characterized by the structure in violet. Ga-radiolabeled annexin V was - as can be seen - also taken up by the liver. The images were obtained two days after infarct and were acquired over one hour.
  • Radionuclides linked to a CD74 antibody as therapeutic agents for B-cell lymphoma comparison of Auger electron emitters with beta-particle emitters. J. Nucl. Med. 41, 2089-2097.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Physics & Mathematics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A thiol-NOTA derivative of Formula (I): wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; and m is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 is described herein. The thiol-NOTA derivatives are suitable for kit 68Ga radiolabeling of proteins and peptides.

Description

TITLE
THIOL-NOTA DERIVATIVES FOR KIT 68Ga
RADIOLABELING
FIELD
[0001] The present disclosure broadly relates to a composition of matter for introducing 68Ga into proteins and peptides. More specifically, but not exclusively, the present disclosure relates to novel 1 ,4,7-triazacyclononane- 1,4,7- triacetic acid (NOTA) derivatives. The present disclosure also relates to Ga radiolabeled l,4,7-triazacyclononane-l,4,7-triacetic acid (NOTA) derivatives. Moreover, the present disclosure also relates to the use of such radiolabeled derivatives for positron emission tomography (PET) imaging.
BACKGROUND
[0002] Commonly used radionuclides for PET imaging include C, F,
18O, 13N, and 76Br. Bioactive molecules labeled with radionuclides suitable for molecular imaging (e.g. PET) purposes are of increasing interest. The radionuclide
Ga has gained in popularity as it enables the fast, simple and convenient labeling of peptides and other small biologically active materials such as antisense oligonucleotides. Furthermore, the labeling with 68Ga is independent from cyclotrons and is thus highly flexible as it can be readily generated from a 68Ge/68Ga generator system.
[0003] Peptides labeled with the PET radionuclide 68Ga via 1,4,7,10- tetraazacyclodocecane-iV,iV',N",N'"-tetraacetic acid (DOTA) exhibit favorable properties for nuclear imaging. In a comparative evaluation study, 68Ga-DOTATOC, a somatostatin analog, was superior to the commonly used n 'in-labeled Octreoscan® for detecting neuroendocrine tumors, largely due to the higher mean and maximum standardized uptake values and higher tumor/non-tumor ratios in lung and skeletal manifestations. In 77.8% of discrepant osseous findings, positive PET interpretations were verified as true [I]. After elution from the Ge/ Ga generator system, 68Ga3+ was mixed with the peptide-chelator-conjugate DOTATOC and kept at pH 4.8 and a temperature of 9O0C over a period of 15 min (conditions that would denature any protein).
[0004] Wei et al. have described the synthesis and use of 68Ga-labeled
MSH in melanoma [2]. The synthesis Of 68Ga labeled DOTA-ReCCMSH(Argl l) involved reactions carried out at room temperature over periods of 3 hours, at 9O0C over periods of 2 hours, and at 850C over periods of 30 minutes at pH 3.8-4. The overall synthetic yield of unlabelled peptide-chelator-conjugate was reported to be 10-20%. Acute biodistribution and PET imaging performed on mice carrying B 16/Fl melanoma tumors showed moderate receptor-mediated tumor uptake, fast non-target organ clearance, high tumor/non-target tissue ratios, and clear visualization at all time points examined up to 2 hours following injection.
[0005] Despite its easier synthesis, 68Ga-labeled RGD did not detect its alphavbeta3 integrin receptors as efficiently as 18F-galacto-RGD and π iIn-labelled RGD [3]. In a common procedure, following the conjugation of the peptide cycloRGDfK with DOTA, the conjugate is deprotected at room temperature over a period exceeding 25 hours. Labeling with Ga occurred within 7 min at 8O0C with a radiochemical purity >95%. Labeling with 111In occurred within 25 min at 95°C with a radiochemical purity >90%. Moreover, the radiochemical yield of 68Ga- DOTA-RGD was shown to be approximately 60% as compared to approximately 30% for 18F-galacto-RGD. While tumor uptake ratios were comparable, tumor/blood ratios were higher for 111In-DOTA-RGD and 18F-galacto-RGD than for 68Ga-DOTA- RGD, largely due to a higher blood pool activity {i.e. higher background activity). [0006] According to Decristoforo et al, the lack of stability of 68Ga
DOTA-peptides could be overcome by using alternative chelator systems such as p- SCN-Bn-NOTA, which could also be better suited for the clinical use of gallium [3]. While the currently used NOTA derivatives are well-suited for the derivatization of peptides, they do not yet allow the derivatization of proteins.
[0007] The cleavage of the
Figure imgf000004_0001
protecting groups of the monoreactive NOTA derived prochelator NODAGA(^BU)3 (4-(4,7-bis(2-tert- butoxy-2-oxoethyl)- 1 ,4,7-triazonan- 1 -yl)-5-ter/-butoxy-5-oxopentanoic acid) requires concentrated trifluoroacetic acid [4]. NODAGA(tBu)3 was synthesized in five steps in an overall yield of 21% and was subsequently coupled to the peptide [Tyr3] -octreotide (TOC) providing the peptide-chelator conjugate NODAGATOC. The conjugate was used as a SPECT and PET tracer when labeled with 111In, 67Ga, or 68Ga.
[0008] The chelator p-SCN-Bn-NOTA (5'-2-(4-isothiocyanatobenzyl)- l,4,7-triazacyclononane-l,4,7-triacetic acid) was shown to achieve only moderate yields of the corresponding chelator-protein conjugates [5]. Peptide-chelator conjugates, NOTA-RGD, were prepared under standard SCN-amine reaction condition {i.e. stirring at room temperature over a period of 5 hours and isolation by semi-preparative HPLC) producing NOTA-RGDl (NOTA-c(RGDyK)) in 61% yield, NOTA-RGD2 (NOTA-E[c(RGDyK)]2) in 52 % yield, and N0TA-RGD4 (NOTA-E {E[c(RGDyK)]2}2) in 43% yield. The 68Ga labeling procedure was conducted using 185 MBq Of 68Ga over a period of 10 minutes at 40°C. Radioactive Ga-NOTA-RGD peptides were purified by semi-preparative HPLC leading to decay-corrected yields of 90% for NOTA-RGDl, 82% for NOTA-RGD2 and 64% for N0TA-RGD4. The specific activity of the 68Ga-NOTA-RGD multimers was measured to be about 9.7-13.6 MBq/nmol. Similar results have been published [6]. [0009] Velikyan et al. have previously reported a straightforward labeling method for quickly introducing macrocyclic bifunctional chelators into Ga peptide-chelator conjugates at room temperature as demonstrated for NODAGA- TATE in which NOTA has been coupled to the eight amino acid residue peptide [Tyr3]Octreotate [7]. Formation kinetics of 68Ga-NOTA, studied as a function of pH, and formation kinetics of 68Ga-NODAGA-TATE, studied as a function of the bioconjugate concentration, indicated a nearly quantitative radioactivity incorporation of more than 95% for 68Ga-NOTA within less than 10 minutes at room temperature and at pH 3.5, and from 90-95% for 68Ga-NODAGA-TATE respectively. The purification of the 68Ga-labeled products was described as being unnecessary since the radiochemical purity was >95%. Additional NOTA derivatives, such as TACN-TM (l,4,7-tris(2-mercaptoethyl)-l,4,7- triazacyclononane), NODASA (1,4,7-tri-azacyclononane-l -succinic acid-4,7- deacetic acid), NOTP (l,4,7-triazacyclononane-N,N',N"- tris(methylenephosphonic)acid), and NOTPME (l,4,7-triazacyclononane-N,N',N"- tris(methyl-enephosphonate-monoethylester)) were shown to demonstrate high thermodynamic stability and similar plasma and in vivo stability. While the authors suggest this method to be suitable for 68Ga-labelling of proteins, the allegation has not been substantiated since the NOTA-NHS-ester can only be introduced into macromolecules at pH values of 8.5-9 and using incubation periods of several hours. This conjugation step would adversely affect the NHS-ester of NOTA itself by leading to a high rate of hydrolysis, as it would most proteins [8, 9].
[0010] Only one method has thus far been described for introducing
68Ga into antibody fragments [10]. Although N,N'-bis[2-hydroxy-5- (carboxyethyl)benzyl]ethylene-di-amine-N,N'-diacetic acid (HBED-CC)-derivatized antibodies could be generated, the labeling efficiency and product yield dropped with increasing reaction temperature. Lower reaction temperatures however, entailed the requirement for purification steps. Moreover, the chelator is only accessible following a complex multi-step synthesis. [0011] Contrary to 68Ga, ^-SCN-Bn-NOTA was reported as being successfully applied for introducing 67Ga (an Auger electron emitter) into antibodies as well as into an antibody fragment [11, 12, 13, 14]. Although it allowed for introducing 67Ga into protein molecules, p-SCN-Bn-NOTA only provided for low radiochemical yields for 68Ga, making subsequent purification steps necessary. Moreover, reaction times ranging from 30-60 minutes are incompatible with the short half-life Of68Ga.
[0012] The p-SCN-Bn-NOTA chelator has an additional inherent disadvantage. While only a limited number of chelators can be introduced into peptides, proteins might have more than one site capable of accepting a chelator. While an increased number of chelators per macromolecule could increase the specific activity of the chelator-protein conjugate, it has been shown that a high number of derivatization sites leads to a dramatic loss of the biological activity of the derivatized biomolecule [15, 16, 17]. In case of the isothiocyanate derivative of NOTA, the number of derivatization sites per protein molecule could only be determined by means of 14C labeled chelators or isotopic dilution titration which represent highly complex methods.
[0013] The present disclosure refers to a number of documents, the content of which is herein incorporated by reference in their entirety.
SUMMARY
[0014] The present disclosure broadly relates to thiol-NOTA derivatives.
[0015] As broadly claimed, the present disclosure relates to a thiol-
NOTA derivative of Formula I:
Figure imgf000007_0001
[0016] wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; and
[0017] m is 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10.
[0018] In an embodiment, the present disclosure relates to a thiol-
NOTA derivative of Formula Ia:
Figure imgf000007_0002
[0019] In an embodiment, the present disclosure broadly relates to Ga radiolabeled thiol-NOTA derivatives.
[0020] As broadly claimed, the present disclosure relates to a 68Ga radiolabeled thiol-NOTA derivative of Formula II:
Figure imgf000008_0001
[0021] wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; and
[0022] m is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
[0023] In an embodiment, the present disclosure relates to a 68 G/ a radiolab led thiol-NOTA derivative of Formula Ha:
Figure imgf000008_0002
[0024] In a further embodiment, the present disclosure broadly relates to thiol-NOTA derivatives for protein and peptide labeling.
[0025] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for protein and/or peptide labeling.
[0026] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for protein labeling. [0027] In yet a further embodiment, the present disclosure relates to the se of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling hMAb 425.
[0028] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling human serum albumin (HSA).
[0029] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling rat serum albumin (RSA).
[0030] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling bovine serum albumin (BSA).
[0031] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling diphtheria toxin (CRM mutant).
[0032] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula Ha for labeling human annexin V.
[0033] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia. [0034] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and hMAb 425.
[0035] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and human serum albumin (HSA).
[0036] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and rat serum albumin (RSA).
[0037] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and bovine serum albumin (BSA).
[0038] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and diphtheria toxin (CRM mutant).
[0039] In yet a further embodiment, the present disclosure relates to thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula I or Formula Ia and human annexin V.
[0040] In yet a further embodiment, the present disclosure relates to
68GGaa rraaddiioollaabbeelleedd tthhiiooll--NNOOTTAA--pprrootteeiin conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha. [0041] In yet a further embodiment, the present disclosure relates to
68Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and hMAb 425.
[0042] In yet a further embodiment, the present disclosure relates to
68Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and human serum albumin (HSA).
[0043] In yet a further embodiment, the present disclosure relates to
68Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and rat serum albumin (RSA).
[0044] In yet a further embodiment, the present disclosure relates to
Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula Ha and bovine serum albumin (BSA).
[0045] In yet a further embodiment, the present disclosure relates to
Aft
Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and diphtheria toxin (CRM mutant).
[0046] In yet a further embodiment, the present disclosure relates to
Ga radiolabeled thiol-NOTA-protein conjugates comprising a thiol-NOTA derivative of Formula II or Formula IIa and human annexin V.
[0047] In yet a further embodiment, the present disclosure relates to the use of thiol-NOTA derivatives of Formula I, Formula Ia, Formula II and Formula IIa for peptide labeling. [0048] In yet a further embodiment, the present disclosure relates to the use of 68Ga-radiolabelled thiol-NOTA derivatives of Formula II and Formula Ha for positron emission tomography (PET) imaging.
[0049] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit comprising a thiol-NOTA derivative of Formula I or Formula Ia.
[0050] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit comprising a thiol-NOTA derivative of Formula I or Formula Ia and a maleimide-based linker for conjugating the thiol-NOTA derivative of Formula I or Formula Ia to a target protein.
[0051] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and a target protein.
[0052] In yet a further embodiment, the present disclosure relates to a
Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and hMAb 425.
[0053] In yet a further embodiment, the present disclosure relates to a
Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and human serum albumin (HSA).
[0054] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and rat serum albumin (RSA). [0055] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and bovine serum albumin (BSA).
[0056] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and diphtheria toxin (CRM mutant).
[0057] In yet a further embodiment, the present disclosure relates to a
68Ga protein labeling kit including a thiol-NOTA-protein conjugate comprising a thiol-NOTA derivative of Formula I or Formula Ia and human annexin V.
[0058] In yet a further embodiment, the present disclosure relates to a method for 68Ga radiolabeling of a target protein, the method comprising:
[0059] reacting the target protein with a maleimide containing reagent to afford a maleimide-modified target protein;
[0060] reacting the maleimide modified protein with a thiol-NOTA derivative of Formula I or Formula Ia to afford a thiol-NOTA-protein conjugate; and
[0061] reacting the thiol-NOTA-protein conjugate with a source of Ga to afford a Ga-radiolabeled thiol-NOTA-protein conjugate.
[0062] In yet a further embodiment, the present disclosure relates to a method for Ga radiolabeling of a target protein, the method comprising: [0063] reacting a thiol-NOTA derivative of Formula I or Formula Ia with a maleimide containing reagent to afford a maleimide-modified thiol-NOTA conjugate;
[0064] reacting the maleimide-modified thiol-NOTA conjugate with a target protein to provide a thiol-NOTA-protein conjugate; and
[0065] reacting the thiol-NOTA-protein conjugate with a source of 68Ga to afford a 68Ga-radiolabeled thiol-NOTA-protein conjugate.
[0066] The foregoing and other objects, advantages and features of the present disclosure will become more apparent upon reading of the following non- restrictive description of illustrative embodiments thereof, given by way of example only.
BRIEF DESCRIPTION OF THE DRAWINGS
[0067] In the appended drawings:
[0068] FIG. 1 shows HPLC chromatograms illustrating the radioactivity traces for labeling mixtures comprising 68Ga-labeled thiol-NOTA- annexin (A), 68Ga-labeled thiol-NOTA-diphtheria toxin CRM mutant (B) and 68Ga- labeled thiol-NOTA-RSA (C). The 68Ga-labeled conjugates were produced using thiol-NOTA derivative 3 (Formula Ia) and linker 5.
[0069] FIG. 2 shows a maximum intensity projection image of a blood pool scan of a healthy rat with Ga-labeled thiol-NOTA-RSA acquired over 30 min, demonstrating radiotracer accumulation in large arteries, the liver, and the heart. Atria and ventricles can be easily identified. RA, right atrium; LA, left atrium; RV, right ventricle; LV, left ventricle; RK, right kidney; LK, left kidney; B, bladder. [0070] FIG. 3 shows a maximum intensity projection image of an anterior myocardial infarction in a rat with [18F] Fluorodeoxyglucose (FDG) (leftside image) and with 68Ga-labeled thiol-NOTA-Annexin V acquired over 60 min (right-side image). The middle image is an overlay of the left and right-side images. The right-side image demonstrates the superiority of direct labeling of apoptotic tissue via apoptosis specific Annexin-V versus negative demonstration by the use of FDG which shows no uptake into apoptotic/necrotic tissue.
DETAILED DESCRIPTION
[0071] In order to provide a clear and consistent understanding of the terms used in the present specification, a number of definitions are provided below. Moreover, unless defined otherwise, all technical and scientific terms as used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention pertains.
[0072] The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one", but it is also consistent with the meaning of "one or more", "at least one", and "one or more than one". Similarly, the word "another" may mean at least a second or more.
[0073] As used in this specification and claim(s), the words
"comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "include" and "includes") or "containing" (and any form of containing, such as "contain" and "contains"), are inclusive or open-ended and do not exclude additional, unrecited elements or process steps. [0074] The term "about" is used to indicate that a value includes an inherent variation of error for the device or the method being employed to determine the value.
[0075] The term "derivative" as used herein, is understood as being a substance similar in structure to another compound but differing in some slight structural detail.
[0076] The present description refers to a number of chemical terms and abbreviations used by those skilled in the art. Nevertheless, definitions of selected terms are provided for clarity and consistency.
[0077] The present disclosure relates to thiol-NOTA derivatives for protein and peptide labeling. In an embodiment, the thiol-NOTA derivatives of the present disclosure function as chelators for Ga.
[0078] In an embodiment of the present disclosure, the thiol-NOTA derivatives of Formula I or Formula Ia are coupled to a target protein using a maleimide-based linker. In a further embodiment of the present disclosure, a target protein is modified using 4-(N-maleimidomethyl)cyclohexane-l-carboxylic acid 3- sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) to produce a maleimide modified protein. Other suitable maleimide-based linkers are known in the art and are within the capacity of a skilled technician. A non-limiting example of such other suitable maleimide-based linkers includes 1 ,2-bis(maleimido)ethane. The maleimide modified proteins exhibited a modified molecular size as well as a different structure when compared to the starting protein. The maleimide modified proteins are subsequently coupled with the thiol-NOTA derivatives of Formula I or Formula Ia. [0079] High protein coupling (i.e. conjugation) efficiencies of the thiol-
NOTA derivatives of Formula I or Formula Ia are important. This observation is further compounded when working with proteins of limited availability and high cost. In an embodiment of the present disclosure, deprotection steps, prior and following protein coupling, are eliminated. Moreover, the number of derivation sites per protein should be controlled such that coupling does not entail loss of biological activity.
[0080] The thiol-NOTA derivatives of the present disclosure afford high coupling rates at ambient temperatures with maleimide modified proteins leading to protein conjugates of high chemical purity. In an embodiment of the present disclosure, the thiol-NOTA-protein conjugates are obtained in yields ranging from 85-99%.
[0081] The maleimide-modified proteins comprise a well-defined number of derivatization sites for coupling with the thiol-NOTA derivatives of Formula I or Formula Ia. The thiol-NOTA protein conjugates (labeling precursors) are readily labeled with Ga to provide Ga-radiolabelled thiol-NOTA-protein conjugates. The radiolabeling process does not affect the biological activity of the target protein. In an embodiment of the present disclosure, the thiol-NOTA-protein conjugates are labeled with Ga within a period of time ranging from about 5 to about 10 minutes. In a further embodiment of the present disclosure, the thiol- NNOOTTAA--pprrootteeiinn ccoonnjjuuggaatteess aarree llaabbeled with 68Ga within a period of time ranging from about 7 to about 10 minutes.
[0082] In an embodiment of the present disclosure, the thiol-NOTA- protein conjugates are labeled with 68Ga in high specific radioactivities ranging from about 10 to about 50 GBq/μmol. In a further embodiment of the present disclosure, the thiol-NOTA-protein conjugates are labeled with 68Ga in radiochemical yields >95%. The 68Ga-radiolabeled thiol-NOTA-protein conjugates of the present disclosure are stable (i.e. radiochemical yields remain substantially constant).
[0083] The synthesis of a thiol-NOTA derivative in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 1. A protected commercially available NOTA-GA(tBu)3 (1) is coupled to S-trityl- mercaptoethanolamine-trifiuoroacetate to provide adduct 2. Subsequent deprotection using concentrated TFA afforded thiol-NOTA derivative 3 (Formula Ia) in moderate overall yield (22%).
Figure imgf000018_0001
Scheme 1
[0084] The synthesis of a thiol-NOTA protein conjugate in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 2. Target protein (4) is derivatized using 4-(N-maleimidomethyl)cyclo-hexane-l- carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) (5) to provide maleimide-modified protein (6). Subsequent conjugation using thiol- NOTA derivative 3 (Formula Ia) provides the thiol-NOTA-protein conjugate labeling precursor (7).
Figure imgf000019_0001
Scheme 2
[0085] The synthesis of a 68Ga-radiolabelled thiol-NOTA protein conjugate in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 3. Thiol-NOTA-protein conjugate (7) is labeled with 68Ga at room temperature and at a pH ranging from about 3.5 to 4 to provide 68Ga-labeled thiol-NOTA-protein conjugate (8).
Figure imgf000019_0002
7 8
Scheme 3
[0086] The synthesis of a maleimide modified thiol-NOTA derivative in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 4.
Figure imgf000020_0001
Scheme 4
[0087] The synthesis of a 68Ga-radiolabelled thiol-NOTA protein conjugate in accordance with an embodiment of the present disclosure is illustrated hereinbelow in Scheme 5.
Figure imgf000020_0002
Scheme 5
[0088] GENERATION OF MALEIMIDE-MODIFIED PROTEINS
[0089] Prior to coupling the thiol-NOTA derivatives of Formula I or
Formula Ia, the target protein was modified using 4-(N- maleimidomethyl)cyclohexane- 1 -carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) to produce a maleimide modified protein. The maleimide group provides for covalent thiol-maleimide coupling at ambient temperatures within a period of time ranging from about 5 to about 10 minutes. The number of maleimide groups introduced into the target protein for subsequent thiol- NOTA coupling could be quantified using Ellman's assay. Target proteins (e.g. annexin, diphtheria toxin (CRM mutant) and BSA) were subjected to a seven-fold excess of 4-(N-maleimidomethyl)cyclohexane-l-carboxylic acid 3-sulfo-N- hydroxysuccinimide ester sodium salt (Sulfo-SMCC) affording maleimide modified proteins comprising from about 1.4 to about 1.7 maleimide groups/protein. This results in only minor alterations of the biological activity of the target protein [15, 16, 17]. The maleimide-modified proteins were purified using size-exclusion gel chromatography (recovery rates of 95-98%).
[0090] The modification of recombinant target proteins with free cysteine residues provides an additional improvement. The introduction of cysteine residues into the target protein, at positions which are not essential for its biological function, leads to an optimization between labeling and optimal biological activity, determining the diagnostic or therapeutic value of a 68Ga labeled radiochemical. The newly introduced cysteine residue(s) allow(s) for the preparation of a maleimide modified protein with a site specific instead of a limited but random modification site.
[0091] GENERATION OF THIOL-NOTA-PROTEIN-
CONJUGATES
[0092] The maleimide-modified proteins were coupled with a thiol-
NOTA derivative of Formula I or Formula Ia in an aqueous medium at pH 7.2 over a period of 30 minutes. Purification by ultrafiltration afforded the labeling precursors (thiol-NOTA-protein conjugates) at yields ranging from 85 to 95% (Scheme 2). Maleimide-modified proteins were generated based on human Monoclonal Antibody 425 (IgG, MW 150 IcDa), RSA (MW 69 kDa), HSA (MW 68 kDa), BSA (MW 66 kDa), diphtheria toxin (CRM mutant) (MW 63 kDa) and human annexin V (MW 36 kDa).
[0093] LABELING OF THE THIOL-NOTA-PROTEIN-
CONJUGATES WITH 68Ga
[0094] The thiol-NOTA-protein conjugates were radiolabeled with 68Ga by mixing each protein conjugate (6.9 nmol) with 1 mL of Ga (240-340 MBq, eluted from a 6 Ge/68Ga generator) in an acetate-buffered aqueous medium at ambient temperature (pH ranging from 3.5 to 4.0) over a period ranging from 5 to 10 minutes (Scheme 3). All labeling reactions were complete after a period of time ranging from 7-10 minutes affording the corresponding Ga radiolabeled thiol- NOTA-protein-conjugates in suitable radiochemical purities. A completed labeling reaction afforded the 68Ga radiolabeled thiol-NOTA-protein-conjugate in a radiochemical purity of more than 95% (FIG. 1) and specific radioactivities ranging between 10 and 50 GBq/μmol. The high radiochemical purity of the 68Ga- radiolabeled thiol-NOTA-protein-conjugates makes a subsequent purification step redundant Moreover, the thiol-NOTA-protein-conjugates were shown to be stable in aqueous solution for at least 4 weeks at 4°C as they could be labeled with unvarying radiochemical yields. The thiol-NOTA derivative of Formula Ia was demonstrated to be stable for 2 years at room temperature.
[0095] MATERIALS AND METHODS
[0096] GENERAL REAGENTS
[0097] All commercially available chemicals were of analytical grade and used without further purification. Sulfo-SMCC and NODA-GA(tBu)3 were purchased from Pierce (Bonn, Germany) and CheMatech (Dijon, France), respectively. S-trityl-mercaptoethanolamine-trifluoroacetate was synthesised according to published procedures [18].
[0098] An Agilent 1200 analytical and semi -preparative HPLC system was used, together with a Chromolith Performance (RP-18e, 100-4.6 mm, Merck, Germany) and a Chromolith SemiPrep (RP- 18e, 100-10 mm, Merck, Germany) column, respectively. ESI mass spectra were obtained using a Finnigan MAT95Q mass spectrometer. NMR spectra were recorded using a Jeol AS500 spectrometer. Thin-layer chromatography was performed using Polygram SIL G/UV254 TLC plates (Macherey-Nagel, Duren, Germany). Size-exclusion gel chromatography was carried out using NAP-5 or NAP-10 columns (GE Healthcare Bio-Sciences AB, Uppsala, Sweden) and size-exclusion fast protein liquid chromatography (FPLC) was performed using a Superdex 200 10/30 GL column (Amersham Biosciences AB, Uppsala, Sweden). Ultrafiltration was performed using Vivaspin 6 centrifugal concentrators (Sartorius, Gόttingen, Germany) with a MWCO of 10 or 100 kDa.
[0099] SYNTHESIS OF røRr-BUTYL 2.2'-(7-(1-!TERr-BUTOXY- l,5-DIOXO-5-(2-(TRITYLTHIO) ETHYLAMINO)PENTAN-2-YLM.4.7-
TRIAZONANE-1.4-DIYL) DIACETATE (S-TRITYL-TRIS- !TERT-BUTYL- THIOL-NOTA) (2)
[00100] A solution of dicyclohexylcarbodiimide (DCC) (189 mg; 919 μmol) in pyridine (5 mL) was added dropwise to a solution consisting of 4-(4,7- bis(2-tert-butoxy-2-oxoethyl)- 1 ,4,7-triazonan- 1 -yl)-5-tert-butoxy-5-oxopentanoic acid (1, NOTA-GA(tBu)3) (500 mg; 919 μmol) and S-trityl-mercaptoethanolamine- trifluoroacetate (293 mg; 919 μmol) in 40 mL water/MeCN 1 : 1 at room temperature. Following overnight reaction, the solvents were evaporated and the product was purified via semi-preparative HPLC using 0-30% MeCN + 0.1% TFA (R, = 3.9 min). The product was isolated as a white solid (343 mg; 406 μmol; 44%) after lyophilisation. 1H NMR (DMSO-d6) (δ, ppm; J, Hz): 7.35-7.20 (m, 15H); 3.97-3.85 (m, 2H); 3.79-3.65 (m, 3H); 3.41 (t, IH, J3=7.3); 3.16-2.82 (bm, 12H); 2.69-2.65 (m, 2H); 2.19 (t, 2H, J3=7.2); 2.12-2.09 (m, 2H); 1.43+1.40+1.39 (3s, 27H). 13C NMR (DMSO-J6) (δ, ppm; J, Hz): 172.08; 171.82; 169.47; 144.99; 129.62; 128.60; 127.32; 82.19; 81.65; 66.47; 63.91; 55.45; 55.27; 51.61; 50.34; 49.47; 48.82; 46.91; 44.97; 38.02; 32.37; 32.01; 28.34; 28.32; 28.27; 25.96. ESI-MS (m/z) for [M + H]+ (calculated): 816.49 (816.14).
[00101] SYNTHESIS OF MERCAPTOETHYLAMINO-NODA-GA
(THIOL-NOTA) (3)
[00102] S-trityl-tris-tert-butyl-thiol-NOTA (2, 343 mg; 406 μmol) was dissolved in a mixture of TFA (5 mL) and TIS (triisopropylsilane; 200 μL) and reacted for 3 hours at room temperature. The volatile components were evaporated and the product was purified using semi-preparative HPLC with 0-20% MeCN + 0.1% TFA (R, = 2.3 min). The product was isolated as a yellow hardening oil (90 mg; 207 μmol; 51%) after lyophilisation. 1H NMR (DMSO-J6) (δ, ppm; J, Hz): 3.97-3.89 (m, 4H); 3.66-3.63 (m, IH); 3.36 (t, 2H, J3=6.4); 3.31-3.11 (m, 12H); 2.64 (t, 2H, J3=6.4); 2.47 (t, 2H, J3=7.7); 2.19-2.01 (m, 2H); 1.29 (s, IH). 13C NMR (DMSO-J6) (δ, ppm; J, Hz): 175.56; 175.35; 175.22; 63.94; 55.62; 50.97; 49.86; 49.84; 49.71; 49.38; 45.97; 45.92; 42.23; 32.78; 24.52; 23.38. ESI-MS (m/z) for [M + H]+ (calculated): 435.2 (435.5).
[00103] GENERAL PROCEDURE FOR THE MALEIMIDE
MODIFICATION OF THIOL-NOTA DERIVATIVES: PREPARATION OF 2,2'-(7-α-CARBOXY-4-(2-α-(2-(2.5-DIOXO-2.5-DIHYDRO-lH-PYRROL-l- YLVETHYL)-2.5-DIOXOPYRROLIDIN-3-YLTHIO)ETHYLAMINOV4- OXO-BUTYLyIAy-TRIAZONANE-M-DIYL)DIACETIC ACID [00104] A solution of 1,1 '-(ethane- l,2-diyl)bis(l//-pyrrole-2,5-dione)
(4.7 mg; 21.6 μmol) in phosphate buffer (0.1M, pH 6.0):MeCN 1 :1 (1 mL) was added to thiol-NOTA 3 and the pH of the solution adjusted to 7.2 by the addition of phosphate buffer (0.5 M, pH 7.2, 200 μL). After a period of 5 minutes, the reaction was complete and the crude product purified by semi-preparative HPLC using 0- 30% MeCN + 0.1% TFA (R, = 3.4 min). The product was isolated as a lightly yellow solid (11.9 mg, 18.1 μmol; 84%) after lyophilisation. 1H NMR (DMSO-J6) (S, ppm; J, Hz): 7.64 (bs, IH); 6.88 (s, 2H); 4.14 (s, IH); 4.01-3.98 (m, IH); 3.93- 3.82 (m, 2H); 3.75-3.61 (m, 4H); 3.54-3.13 (m, 16H); 3.06-2.98 (m, IH); 2.85-2.77 (m, IH); 2.52-2.48 (m, 2H); 2.46-2.40 (m, 2H); 2.23-2.08 (m, 2H). 13C NMR (DMSO-J6) (S, ppm; J, Hz): 177.76; 175.52; 173.80; 171.67; 171.67; 160.03; 135.28; 64.01; 55.89 + 55.85 + 55.76 + 55.48 + 55.41 + 52.12 + 52.07 + 52.04 + 51.19 + 51.04 + 50.84 + 50.76 + 49.23 + 49.13 + 49.11 + 47.65 + 47.49 + 47.45 + 47.33 + 45.92 + 45.80 + 45.57; 39.86; 39.23; 39.11; 36.71 ; 35.95; 33.05; 32.15; 26.03. ESI-MS (m/z) for [M + H]+ (calculated): 656.0 (655.7). MALDI-MS (m/z) for [M + H]+ (calculated): 656.0 (655.7).
[00105] GENERAL PROCEDURE FOR THE MALEIMIDE
MODIFICATION OF PROTEINS WITH SULFO-SMCC
[00106] A freshly prepared solution of sulfo-SMCC (4-(N- maleimidomethyl)cyclohexane- 1 -carboxylic acid 3 -sulfo-N-hydroxysuccinimide ester sodium salt) (92μg; 210 nmol) in 150 μL of DMF:water (1 :1) was added to a solution of the target protein (30 nmol) in 500 μL PB (0.1 M, pH 7.2). After one hour of incubation, the maleimide-modified protein was purified via size-exclusion chromatography using NAP-5 columns. The maleimide-modified proteins were obtained in high yields (85-99%). The number of maleimide groups per protein was determined using MESNA and Ellman's assay and was found to be ranging between 1.4 and 1.7. [00107] GENERAL PROCEDURE FOR GENERATING THIOL-
NOTA-PROTEIN CONJUGATES
[00108] A freshly prepared solution of thiol-NOTA (3, 131 μg; 300 nmol) was added to a solution comprising a maleimide-modified protein in phosphate buffer (0.1M, pH 7.2) and stirred over a period of 30 minutes. The proteinaceous solution was subsequently diluted with HEPES buffer (0.025 M, pH 4.0) and purified by ultrafiltration applying Vivaspin 6 centrifugal concentrators (MWCO 10 IcDa for annexin V, HSA, BSA, RSA and diphtheria toxin (CRM mutant); MWCO 100 kDa for hMAb 425). The ultrafiltration was repeated four times. The thiol-NOTA-protein conjugates were recovered at high yields (85-99%).
[00109] GENERAL PROCEDURE FOR THE COUPLING OF
MALEIMIDE-MODIFIED THIOL-NOTA DERIVATIVES WITH THIOL CONTAINING PROTEINS
[00110] To a solution of SDF-I α [chemokine stromal-derived factor- lα; a small protein (MW HkDa) shown to accumulate in mycard infarct tissue] (1 mg, 91 nmol) in phosphate buffer (0.1 M, pH 6.0, 500 μL) was added a solution of TCEP (Tris(2-Carboxyethyl) phosphine Hydrochloride) (5 eq., 455 nmol, 0.13 mg) in water (100 mL). The resulting mixture was reacted over a period of 10 minutes followed by the addition of a solution comprising NOTA-maleimide (10 eq., 909 nmol, 0.6 mg) in phosphate buffer (0.1 M, pH 6.0, 200 μL) (Scheme 5). The pH of the solution was adjusted to 7.2 by the addition of phosphate buffer (0.5 M, pH 7.2, 50 μL). After 30 minutes, the crude NOTA-protein-conjugate was purified by semi- preparative HPLC using 0-50% MeCN + 0.1% TFA. The product was isolated as a white solid (1.02 mg, 89 nmol, 98%) after lyophilisation. Radiolabeling was carried out as described hereinbelow. [00111] LABELING OF THIOL-NOTA-PROTEIN-CONJUGATES
WITH 68Ga
[00112] A solution of a thiol-NOTA-protein conjugate (6.9 nmol) in
HEPES buffer (0.025M, pH 4.0) was added to 240-340 MBq of 68Ga in a sodium acetate solution (1 mL; 0.11 M; pH 3.5-4.0) and incubated for 5 to 10 minutes. The reaction mixtures were subsequently analyzed by analytical HPLC. The Ga- radiolabeled thiol-NOTA-protein conjugates were found to be 95-99% pure and obtained in specific radioactivities ranging from 10 and 50 GBq/μmol.
[00113] IN VIVO BIODISTRIBUTION OF 68Ga RADIOLABELED
THIOL-NOTA-RSA IN HEALTHY RATS
[00114] CD rats were housed according to German animal protection laws and protocols of the local committee. Experiments were performed under inhalation anesthesia (oxygen 1.2 L/min, isofluran 1.5 vol %) and appropriate warming to prevent hypothermia.
[00115] 20 to 60 MBq of 68Ga radiolabeled thiol-NOTA-RSA were intravenously administered via the lateral tail vein of CD rats anesthetized by inhalation (oxygen 1.2 L/min, isofluran 1.5 vol %). List mode data were acquired over 30 minutes using a Siemens Inveon 120 small animal PET system (Siemens Preclinical Imaging, Knoxville, TN) and divided into temporal frames of increasing length varying from 20 s to 5 min for the assessment of temporal changes in regional tracer accumulation. Tomographic volumes were created using 3D iterative reconstruction, yielding a transaxial spatial resolution of approximately 1.4 mm. Rat biodistribution data showed nearly constant whole blood radioactivity from minute 2 over the next 25 min after injection. In previous experiments with 18F-labeled RSA, it was demonstrated that its uptake by the liver was as high as in the blood pool and its uptake by the kidneys in a range similar to the arterial blood volume [19]. In contrast to these results, Ga labeled thiol-NOTA-RSA showed a lower uptake especially in the liver in the first 60 minutes following tracer administration. This can be attributed to a lower lipophilicity of the Ga-labeled albumin resulting from a derivatization leading to the less lipophilic derivatization..
[00116] As shown in FIG. 2, 68Ga-radiolabeled thiol-NOTA-RSA provides a blood pool image of a healthy rat including a clear illustration of the heart, aorta, arteries, and the liver. In addition, radioactivity accumulation can be seen in the kidneys and the bladder due to metabolization and excretion processes.
[00117] IN VIVO BIODISTRIBUTION OF 68GA RADIOLABELED
THIOL-NOTA-ANNEXIN V IN A RAT INFARCT MODEL
[00118] 30-40 MBq of 68Ga-radiolabeled annexin V were intravenously administered via the lateral tail vein of the rats anesthetized by inhalation (oxygen 1.2 L/min, isofiuran 1.5 vol %). List mode data were acquired over 60 minutes using a Siemens Inveon 120 small animal PET system (Siemens Preclinical Imaging, Knoxville, TN) and divided into temporal frames of 10 min for the assessment of temporal changes in regional tracer accumulation. Tomographic volumes were created using 3D iterative reconstruction, yielding a transaxial spatial resolution of approximately 1.4 mm.
[00119] The left coronary artery (LCA) was ligated to induce transmural myocardial infarction. Under anesthesia with intramuscular administration of midazolam (0.1 mg/kg), fentanyl (1 mg/kg), and medetomidin (10 mg/kg) (MMF) and mechanical ventilation, the chest was opened to expose the heart. A 7-0 polypropylene suture on a small curved needle was passed through the LCA and ligated to occlude the LCA. To assess the relationship of in vivo and ex vivo measurements in acute and subacute phases, animals were allowed 2 days of recovery before PET imaging.
[00120] Annexin V was used as an indicator for apoptotic tissue, as can be found in infarcted tissue. While the imaging of a healthy heart with [18F] Fluorodeoxyglucose (FDG) would demonstrate a V-shaped structure, this shape is interrupted in an infarcted heart due to apoptotic tissue that can no longer absorb any nutrients, (i.e. glucose). Contrary to the lack of FDG in the apoptotic infarcted tissue, the use of annexin V as a marker for apoptosis can directly depict the infarcted site.
[00121] FIG. 3 shows the overlay of two maximum intensity projection images of an anterior myocardial infarction in a rat. The image covers a cross section through the coronary level of the heart. Negative landmarking of the infarct region was obtained by the use of [18F] Fluorodeoxyglucose (FDG) while 68Ga- radiolabelded annexin V allows positive infarct imaging. FDG landmarking indicates the infarct region by the lack of accumulating FDG depicted in colors ranging from yellow to red, i.e. the bright apical dotted structure and the interior light and medium grey horizontal C-shaped structure. The direct accumulation of the Ga-radiolabeled annexin V in the apoptotic and necrotic infarct heart tissue is characterized by the structure in violet. Ga-radiolabeled annexin V was - as can be seen - also taken up by the liver. The images were obtained two days after infarct and were acquired over one hour.
[00122] It is to be understood that the invention is not limited in its application to the details of construction and parts as described hereinabove. The invention is capable of other embodiments and of being practiced in various ways. It is also understood that the phraseology or terminology used herein is for the purpose of description and not limitation. Hence, although the present invention has been described hereinabove by way of illustrative embodiments thereof, it can be modified, without departing from the spirit, scope and nature of the subject invention as defined in the appended claims.
REFERENCES
1. Buchmann I, Henze M, Engelbrecht S, Eisenhut M, Runz A, Schafer M, Schilling T, Haufe S, Herrmann T, Haberkorn U. (2007) Comparison of 68Ga- DOTATOC PET and H lIn-DTPAOC (Octreoscan) SPECT in patients with neuroendocrine tumors. Eur J. Nucl. Med. MoI. Imaging. 34, 1617-26.
2. Wei L, Miao Y, Gallazzi F, Quinn TP, Welch MJ, Vavere AL, Lewis JS. (2007) Gallium-68-labeled DOTA-rhenium-cyclized alpha-melanocyte-stimulating hormone analog for imaging of malignant melanoma. Nucl. Med. Biol. 34, 945-53.
3. Decristoforo C, Hernandez Gonzalez I, Carlsen J, Rupprich M, Huisman M, Virgolini I, Wester HJ, Haubner R. (2008) 68Ga- and π 'in-labelled DOTA-RGD peptides for imaging of alphavbeta3 integrin expression. Eur. J. Nucl. Med. MoI. Imaging 35, 1507-1515.
4. Eisenwiener, K.P., Prata, M.I., Buschmann, I., Zhang, H.W., Santos, A.C., Wenger, S., Reubi, J.C., Macke, H.R. (2002) NODAGATOC, a new chelator- coupled somatostatin analogue labeled with 67/68Ga and 111In for SPECT, PET, and targeted therapeutic applications of somatostatin receptor (hsst2) expressing tumors. Bioconj. Chem. 13, 530-541.
5. Li, Z.-B., Chen, K., Chen, X. (2008) 68Ga-labeled multimeric RGD peptides for microPET imaging of integrin alpha(v)beta (3) expression. Eur. J. Med. MoI. Imaging 35, 1100-1108.
6. Jeong, J. M., Hong, M. K., Chang, Y.S., Lee, Y.S., Kim, Y.J., Cheon, G.J., Lee, D. S., Chung, J.K., Lee, M. C. (2008) Preparation of a promising angiogenesis PET imaging agent: Ga-labeled c(RGDyK)-isothiocyanatobenzyl- 1,4,7- triazacyclononane-l,4,7-triacetic acid and feasibility studies in mice. J. Nucl. Med. 49, 830-836.
7. Velikyan, I., Maecke, H., Langstrom, B. (2008) Convenient preparation of Ga- based PET-radiopharmaceuticals at room temperature Bioconj. Chem. 19, 569-573.
8. Cuatrecasas, P., Parikh, I. (1972) Adsorbents for affinity chromatography. Use of N-hydroxysuccinimide Esters of Agarose. Biochemistry 11, 2291-2299.
9. Mier, W., Hoffend, J., Kramer, S., Schuhmacher, J., Hull, W.E., Eisenhut, M., Haberkorn, U. (2005) Conjugation of DOTA using isolated phenolic active esters: the labeling and biodistribution of albumin as blood pool marker. Bioconj. Chem. 16, 237-240.
10. Eder, M., Wangler, B., Knackmuss, S., Legall, F., Little, M., Haberkorn, U., Mier, W., Eisenhut, M. (2008) Tetrafmorophenolate of HBED-CC: a versatile conjugation agent for 68Ga-labeled small recombinant antibodies Eur. J. Nucl. Med. MoI. Imaging 35, 1878-1886. 11. Wu, C, Jagoda, E., Brechbiel, M., Webber, K.O., Pastan, L, Gansow, O., Eckelman, W.C. (1997) Biodistribution and catabolism of Ga-67-labeled anti-Tac dsFv fragment. Bioconj. Chem. 8, 365-369.
12. Lee, J., Garmestani, K., Wu, C, Brechbiel, M.W., Chang, H.K., Choi, C.W., Gansow, O. A., Carrasquillo, J.A., Paik, CH. (1997) In vitro and in vivo evaluation of structure-stability relationship of ' 11In- and 7Ga-labeled antibody via 1 B4M or C- NOTA chelates. Nucl. Med. Biol. 24, 225-230.
13. Govindan, S.V., Goldenberg, D.M., Elsamra, S.E., Griffiths, G.L., Ong, G.L., Brechbiel, M.W., Burton, J., Sgouros, G., Mattes, M.J. (2000) Radionuclides linked to a CD74 antibody as therapeutic agents for B-cell lymphoma: comparison of Auger electron emitters with beta-particle emitters. J. Nucl. Med. 41, 2089-2097.
14. Ochakovskaya, R., Osorio, L., Goldenberg, D.M., Mattes, M.J. (2001) Therapy of disseminated B-cell lymphoma xenografts in severe combined immunodefϊcient mice with an anti-CD74 antibody conjugated with ' ' indium, 67gallium, or 90yttrium. Clin. Cancer Res. 7, 1505-1510.
15. Brechbiel, M. W., Gansow, O. A., Atcher, R. W., Schlom, J., Esteban, J., Simpson, D. E., and Colcher, D. (1986) l-(p-isothiocyanatobenzyl)derivatives of DTPA and EDTA. Antibody labeling and tumor imaging studies Inorg. Chem. 25, 2772-2781.
16. Nikula, T. K., Boccia, M., Curcio, M. J., Sgouros, G., Ma, Y., Finn, R. D., and Scheinberg, D. A. (1995) Impact of the high tyrosine fraction in complementarity determining regions: measured and predicted effects of radioiodination on IgG immunoreactivity. MoI. Immunol. 32, 865-872.
17. Wangler C, G., Moldenhauer, M. Eisenhut, U. Haberkorn, and W. Mier (2008) Antibody-dendrimer conjugates: the number, not the size of the dendrimers, determines the immunoreactivity. Bioconj. Chem. 19, 813-820.
18. Maltese, M. (2001) Reductive demercuration in deprotection of trityl thioethers, trityl amines, and trityl ethers. J. Org. Chem. 66, 7615-7625.

Claims

WHAT IS CLAIMED IS;
1. A thiol-NOTA derivative of Formula I:
Figure imgf000033_0001
wherein: n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; and m is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
2. The thiol-NOTA derivative as defined in claim 1, wherein n and m are 2.
3. A 68Ga radiolabeled thiol-NOTA derivative of Formula II:
Figure imgf000033_0002
wherein: n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; and m is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
4. The thiol-NOTA derivative as defined in claim 3, wherein n and m are 2.
5. Use of the thiol-NOTA derivative as defined in claims 1 or 2 for protein or peptide labelling.
6. The use as defined in claim 5, wherein the proteins are selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
7. Use of the thiol-NOTA derivative as defined in claims 3 or 4 for protein or peptide labelling.
8. The use as defined in claim 7, wherein the proteins are selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
9. A thiol-NOTA protein conjugate comprising a thiol-NOTA derivative as defined in claims 1 or 2 and a target protein.
10. The thiol-NOTA protein conjugate as defined in claim 9, wherein the target protein is selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
11. A 68Ga radiolab led thiol-NOTA protein conjugate comprising a thiol-NOTA derivative as defined in claims 3 or 4 and a target protein.
12. The thiol-NOTA protein conjugate as defined in claim 11, wherein the target protein is selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
13. A Ga protein labeling kit comprising a thiol-NOTA derivative as defined in claims 1 or 2.
14. The 68Ga protein labeling kit as defined in claim 13, further comprising and a maleimide-based linker for conjugating the thiol-NOTA derivative to a target protein.
15. The 68Ga protein labeling kit as defined in claim 14, wherein the target protein is selected from the group consisting of consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
16. A Ga protein labeling kit comprising a thiol-NOTA-protein conjugate, wherein the conjugate is obtained by coupling of a maleimide-modified protein and a thiol-NOTA derivative as defined in claims 1 or 2.
17. The Ga protein labeling kit as defined in claim 16, wherein the maleimide-modified protein is selected from the group consisting of consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
18. A method for Ga radiolabeling of a target protein, the method comprising: a) reacting the target protein with a maleimide containing reagent to afford a maleimide-modified target protein; b) reacting the maleimide modified protein with a thiol-NOTA derivative as defined in claims 1 or 2 to afford a thiol-NOTA- protein conjugate; and c) reacting the thiol-NOTA-protein conjugate with a source of 68Ga to afford a 68Ga-radiolabeled thiol-NOTA-protein conjugate.
19. The method as defined in claim 18, wherein the target protein is selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
20. A method for 68Ga radiolabeling of a target protein, the method comprising: a) reacting a thiol-NOTA derivative as defined in claims 1 or 2 with a maleimide containing reagent to afford a maleimide-modified thiol-NOTA conjugate; b) reacting the maleimide-modified thiol-NOTA conjugate with a target protein to provide a thiol-NOTA-protein conjugate; and c) reacting the thiol-NOTA-protein conjugate with a source of Ga to afford a 68Ga-radiolabeled thiol-NOTA-protein conjugate.
21. The method as defined in claim 20, wherein the target protein is selected from the group consisting of hMAb 425, HSA, RSA, BSA, diphtheria toxin (CRM mutant), and human annexin V.
PCT/CA2009/001822 2008-12-12 2009-12-14 Thiol-nota derivatives for kit 68ga radiolabeling WO2010066051A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12223208P 2008-12-12 2008-12-12
US61/122,232 2008-12-12

Publications (1)

Publication Number Publication Date
WO2010066051A1 true WO2010066051A1 (en) 2010-06-17

Family

ID=42242281

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2009/001822 WO2010066051A1 (en) 2008-12-12 2009-12-14 Thiol-nota derivatives for kit 68ga radiolabeling

Country Status (1)

Country Link
WO (1) WO2010066051A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011033112A3 (en) * 2009-09-21 2011-06-30 Ge Healthcare Limited 68ga-labeled human serum albumin
WO2013086396A1 (en) * 2011-12-09 2013-06-13 Ikaria Development Subsidiary One Llc Labeled alginate conjugates for molecular imaging applications

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000061195A1 (en) * 1999-04-14 2000-10-19 Diatide, Inc. Benzodiazepine derivatives for imaging thrombi
CA2625196A1 (en) * 2005-10-07 2007-04-19 Guerbet Compounds comprising a biological target recognizing part, coupled to a signal part capable of complexing gallium

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000061195A1 (en) * 1999-04-14 2000-10-19 Diatide, Inc. Benzodiazepine derivatives for imaging thrombi
CA2625196A1 (en) * 2005-10-07 2007-04-19 Guerbet Compounds comprising a biological target recognizing part, coupled to a signal part capable of complexing gallium

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WELCH, M. J. ET AL.: "A Novel Hexachelating Amino-Thiol Ligand and Its Complex with Gallium (III)", INORG. CHEM., vol. 29, no. 4, 1990, pages 672 - 676 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011033112A3 (en) * 2009-09-21 2011-06-30 Ge Healthcare Limited 68ga-labeled human serum albumin
WO2013086396A1 (en) * 2011-12-09 2013-06-13 Ikaria Development Subsidiary One Llc Labeled alginate conjugates for molecular imaging applications
US8974766B2 (en) 2011-12-09 2015-03-10 Bellerophon Bcm Llc Labeled alginate conjugates for molecular imaging applications
US9260399B2 (en) 2011-12-09 2016-02-16 Bellerophon Bcm Llc Labeled alginate conjugates for molecular imaging applications
US9562113B2 (en) 2011-12-09 2017-02-07 Bellerophon Bcm Llc Labeled alginate conjugates for molecular imaging applications

Similar Documents

Publication Publication Date Title
CN108541302B (en) Urea-based Prostate Specific Membrane Antigen (PSMA) inhibitors for imaging and therapy
JP2022159345A (en) Label inhibitor of prostate-specific membrane antigen (psma), and application thereof as imaging agent and pharmaceutical for treatment of prostate cancer
ES2749640T3 (en) Radiolabeled octreota analogs as PET tracers
Bandari et al. Synthesis and biological evaluation of copper-64 radiolabeled [DUPA-6-Ahx-(NODAGA)-5-Ava-BBN (7-14) NH2], a novel bivalent targeting vector having affinity for two distinct biomarkers (GRPr/PSMA) of prostate cancer
Kumar et al. In vitro and in vivo evaluation of 64Cu-radiolabeled KCCYSL peptides for targeting epidermal growth factor receptor-2 in breast carcinomas
ES2308778T3 (en) COMPOSITIONS OF RADIOMARCED PEPTIDES TO DIRECT TO A SPECIFIC SITE.
Lindner et al. PESIN multimerization improves receptor avidities and in vivo tumor targeting properties to GRPR-overexpressing tumors
JP7412487B2 (en) Methods and kits for preparing radionuclide complexes
JP2014503546A (en) Radiolabeled HER2 binding peptide
JP2012512233A (en) PDGF-Rβ binding agent
JP6792875B2 (en) HBED-bisphosphonates, their radioactive metal conjugates, and their use as seranostics
Meckel et al. A DOTA based bisphosphonate with an albumin binding moiety for delayed body clearance for bone targeting
JP2014503547A (en) HER2-binding peptide labeled with an organosilicon compound containing 18F
Wu et al. New 68Ga-PhenA bisphosphonates as potential bone imaging agents
Lodhi et al. Development of 99mTc-Labeled human serum albumin with prolonged circulation by chelate-then-click approach: A Potential Blood Pool Imaging Agent
WO2010066051A1 (en) Thiol-nota derivatives for kit 68ga radiolabeling
AU2175492A (en) Method for detecting and localizing tissues having neurokinine 1 receptors
CA2464002C (en) Pacap compositions and methods for tumor imaging and therapy
Grus et al. DOTA conjugate of bisphosphonate and PSMA-inhibitor: A promising combination for therapy of prostate cancer related bone metastases
US6395255B1 (en) Radiolabeled vasoactive intestinal peptide analogs for imaging and therapy
Yang et al. Synthesis and biological evaluation of 99mTc-DMP-NGA as a novel hepatic asialoglycoprotein receptor imaging agent
Suzuki et al. Synthesis and evaluation of a para-carboxylated benzyl-DOTA for labeling peptides and polypeptides
EP4338757A1 (en) New pet tracer for imaging of the functional liver reserve
Braz Mitochondria-Targeted 111In-Radiocomplexes for Auger Therapy of Prostate Cancer
Chandra et al. Synthesis, radiolabeling and biological evaluation of a neutral tripeptide and its derivatives for potential nuclear medicine applications

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: 09831360

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: 09831360

Country of ref document: EP

Kind code of ref document: A1