WO2024110329A1 - Labeling of biomolecules with actinium-225 and medical uses thereof - Google Patents

Labeling of biomolecules with actinium-225 and medical uses thereof Download PDF

Info

Publication number
WO2024110329A1
WO2024110329A1 PCT/EP2023/082190 EP2023082190W WO2024110329A1 WO 2024110329 A1 WO2024110329 A1 WO 2024110329A1 EP 2023082190 W EP2023082190 W EP 2023082190W WO 2024110329 A1 WO2024110329 A1 WO 2024110329A1
Authority
WO
WIPO (PCT)
Prior art keywords
precursor
dota
labeled
psma
freeze
Prior art date
Application number
PCT/EP2023/082190
Other languages
French (fr)
Inventor
Calogero D'ALESSANDRIA
Alexander WURZER
Mathias EIBER
Wolfgang Weber
Original Assignee
Technische Universität München
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 Technische Universität München filed Critical Technische Universität München
Publication of WO2024110329A1 publication Critical patent/WO2024110329A1/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
    • A61K51/0497Organic compounds conjugates with a carrier being an organic compounds
    • 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
    • A61K51/0402Organic compounds carboxylic acid carriers, fatty acids
    • 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
    • A61K51/083Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins the peptide being octreotide or a somatostatin-receptor-binding peptide
    • 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
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/121Solutions, i.e. homogeneous liquid formulation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a method for radioactive labeling of a precursor with 225 Ac comprising freeze-drying the precursor before labeling.
  • the present invention further relates to the 225 Ac-labeled precursor obtained in said method for use in the treatment of cancer.
  • the present invention also relates to a kit for radioactive labeling of a precursor with 225 Ac.
  • Beta radiation is a particulate radiation consisting of high-speed electrons, which lose their kinetic energy, while their track paths become increasingly contorted due to scattering.
  • An exemplary 2 MeV beta particle has a rather long range of 1 cm in water.
  • the linear energy transfer (LET) which represents the total amount of energy deposited per unit track length, is comparably low for beta particles with values around 0.2 keV/pm (Boyd et al., 2006).
  • Alpha particles are positively charged with a mass and charge equal to a helium nucleus. Their energies are higher compared to beta particles with values of 5-9 MeV, displaying a linear track length of 20 to 80 pm in water. Moreover, their higher LET (approx. 100 keV/pm) can result in more lethal double strand DNA breaks, compared to beta particles. In combination with the short range of alphas, the high energy discharge results in a very toxic effect to a relatively small field with very limited damage to surrounding normal tissue (Scheinberg et al., 2011).
  • the alpha emitter Actinium-225 ( 225 Ac) was suggested as a radionuclide for targeted alpha therapy (TAT) of cancer (Morgenstern et al, 2018).
  • TAT targeted alpha therapy
  • 225 Ac is complexed by a suitable chelator, connected to a biological active molecule.
  • the radiopharmaceutical binds to cancer cells and delivers the payload, resulting in irradiation of cancer cells, while minimizing damage to healthy tissues.
  • 225 Ac (ti/2 9.9 d; 6 MeV alpha-particle) decay yields 4 alpha and 3 beta disintegrations (Robertson et al., 2018).
  • the daughter nuclides with suitable gamma emissions, i.e. 221 Fr and 213 Bi, can be exploited for imaging or measuring drug distribution.
  • the decay chain of 225 Ac is summarized below and depicted in Figure 1 :
  • 225 Ac needs to be bound kinetically inert to a chelator and labeling procedures with respective chelators need to be reliable and quantitative.
  • free 225 Ac after incomplete labeling reactions can also be separated by a cartridge-based purification step, but due to the high costs and limited availability of this radionuclide this approach is very uneconomic.
  • chelator for complexation of 225 Ac is the 12-membered macrocyclic ligand 1,4,7, 10-Tetraazacyclododecane-l,4,7,10-tetrayltetraacetic acid (DOTA) or the glutamic-acid substituted derivative (DOTA-GA) (Thiele and Wilson, 2018).
  • DOA 12-membered macrocyclic ligand 1,4,7, 10-Tetraazacyclododecane-l,4,7,10-tetrayltetraacetic acid
  • DOTA-GA glutamic-acid substituted derivative
  • There are numerous 225 Ac-DOTA constructs which have been evaluated in preclinical studies and are also under evaluation in several clinical trials, e.g. for the treatment of leukemia (Jurcic etal., 2017), multiple myeloma, and prostate cancer (Sathekge et al., 2021).
  • DOTA-based chelators are not regarded as ideal chelators for complexation of 225 Ac, because the metal ion radius is too large for the cavity of this chelator (Thiele and Wilson, 2018). Even though, the in vivo stability is considered to be sufficiently high for in vivo applications, a main challenge of this size mismatch between 225 Ac and DOTA, results in difficulties during radiolabeling. In general, complexation of DOTA with 225 Ac requires harsh reaction conditions, including elevated temperatures, micro-wave assistance and long time periods (Thiele and Wilson, 2018). Noteworthy, much research effort is currently dedicated to the development of novel chelators, which allow complexation of 225 Ac under mild reaction conditions. This resulted for example in identification of the larger MACROPA chelator, allowing radiolabeling at room temperature within 5 minutes (Thiele et al., 2017).
  • this object is solved by a method for radioactive labeling of a precursor with 225 Ac, said method comprising the steps of
  • this object is solved by the 225 Ac-labeled precursor obtained in a method according to the present invention for use in the treatment of cancer.
  • kits for radioactive labeling of a precursor with 225 Ac comprising
  • the present invention provides a method for radioactive labeling of a precursor with 225 Ac.
  • Said method comprises the steps of:
  • the precursor is preferably a biomolecule comprising a chelator suitable to complex 225 Ac.
  • the chelator is preferably connected to the biomolecule.
  • Suitable chelators are DOTA, DOTA-GA and MACROPA:
  • PubChem CID 135349208.
  • the precursor is a prostate-specific membrane antigen (PSMA)- targeted ligand.
  • PSMA prostate-specific membrane antigen
  • PSMA-targeted ligands are PSMA I&T, or PSMA-617.
  • PSMA I&T Prostate-specific membrane antigen for imaging & therapy
  • ITM Isotope Technologies Kunststoff SE, Germany or Scintomics Molecular, Applied Theranostics Technologies (SCI-att), Germany.
  • Vipivotide tetraxetan is a high potent prostate-specific membrane antigen (PSMA) inhibitor.
  • PSMA-617 is commercially available, such as from Advanced Accelerator Applications (AAA, a Novartis company), France.
  • the precursor is a somatostatin receptor-targeted ligand.
  • Preferred somatostatin receptor-targeted ligands are octreotide or octreotate, more preferably DOTA-octreotide or DOTA-octreotate
  • Octreotide sold under the brand name Sandostatin among others, is an octapeptide that mimics natural somatostatin pharmacologically. It binds predominantly to the somatostatin receptors SSTR2 and SSTR5.
  • DOTA-TOC (DOTATOC, DOTA-octreotide, DOTA-(Tyr 3 )-octreotide, and DOTA(O)- Phe(l)-Tyr(3))octreotide) is a compound containing tyrosine 3 -octreotide, an SSR agonist, and the bifunctional chelator DOTA.
  • Edotreotide is itse International Nonproprietary Name.
  • Octreotate or octreotide acid is a somatostatin analog that is closely related to octreotide. It is a somatostatin receptor peptide agonist.
  • DOTA-TATE (DOTATATE, DOTA-octreotate, oxodotreotide, DOTA-(Tyr 3 )-octreotate, and DOTA-0-Tyr3-Octreotate) is a compound containing tyrosine 3 -octreotate, an SSR agonist, and the bifunctional chelator DOTA (tetraxetan).
  • the precursor is obtained from a commercial provider
  • it has been sterile filtered by the commercial provider.
  • microbiological evaluations have been carried out by the commercial provider, such as bioburden control and/or the content of bacterial endotoxins has been determined.
  • a precursor is provided.
  • the precursor is dissolved in a buffer solution.
  • the buffer solution is a TRIS (tris(hydroxymethyl)aminomethane) buffer solution, preferably 0.1 M TRIS buffer having pH 7 to 9, more preferably 0.1 M TRIS pH 9.
  • the buffer solution is an acetate buffer solution, preferably 0.1 M to IM acetate pH 4 to 6, more preferably 0.1 M acetate pH 5.7.
  • NaOAc sodium acetate
  • the buffer solution is an acetate buffer solution, preferably 0.1 M to IM acetate pH 3 to 6, more preferably 0.1 M acetate pH 3.7.
  • NaOAc sodium acetate buffer having pH 3.7.
  • the buffer solution is a phosphate buffer solution, preferably 0. 1 M to 0.3 M phosphate pH 5 to 6, more preferably 0.2 M phosphate pH 5.7.
  • the buffer solutions can be sterile filtered before and/or after dissolving the precursor.
  • the buffer solution including the dissolved precursor is sterile filtered.
  • step 2 the precursor is freeze-dried. A powder of the freeze-dried precursor is obtained.
  • step 2 the precursor is freeze-dried while it is dissolved in buffer.
  • the powder of the freeze-dried precursor also contains buffer salt(s).
  • the freeze-drying is via lyophilization.
  • the solution is sterile filtered before lyophilization.
  • step 3 the freeze-dried precursor is reconstituted with water.
  • the freeze-dried precursor is reconstituted with water soon after the freeze-drying, such as immediately. In one embodiment, there is a period of time between obtaining the powder of the freeze-dried precursor and reconstituting it for further use. This means the powder of the freeze-dried precursor can be stored before radiolabeling with 225 Ac.
  • the storage is preferably at about -20°C.
  • the storage time depends on the shelf life of the precursor, which can be up to one year.
  • Said period of time can be in a range from hours to months, such as one to two months, but also up to one year.
  • step 4 225 Ac is provided.
  • 225 Ac is provided in 0.04 M to 0.2 M HC1.
  • 225 Ac is provided in 0.1 M HC1.
  • the activity or dosage of 225 Ac is usually in the range from 4 to 12 MBq.
  • step 5 the 225 Ac of step (4) is added to the reconstituted precursor of (3).
  • 8 MBq of 225 Ac is added.
  • the ratio of 225 Ac to reconstituted precursor is preferably 5 to 20 nmol per MBq 225 Ac, preferably 10 nmol per MBq 225 Ac.
  • step 6 heating for a period of time is carried out.
  • the heating is to at least about 75°C.
  • the heating in step (6) is carried out at about 95°C in a microwave reactor and the period of time is from about 5 to 10 min, preferably about 5 min.
  • the heating in step (6) is carried out at about 75°C to about 85°C and the period of time is from about 20 to 30 min, preferably 75°C for about 20 min.
  • Step (7) is carried out at about 75°C to about 85°C and the period of time is from about 20 to 30 min, preferably 75°C for about 20 min.
  • step 7 the 225 Ac-labeled precursor is obtained.
  • Step (7) preferably comprises cooling.
  • the obtained 225 Ac-labeled precursor is formulated, preferably in isotonic saline, and/or sterile filtered.
  • the 225 Ac-labeled precursor obtained in step (7) can be formulated in isotonic saline.
  • the formulation optionally contains one or more stabilizing agents and/or a co-chelator.
  • stabilizing agents examples are:
  • DTP A diethylenetriamine pentaacetate
  • Said co-chelator is capable of chelating radioactive daughter nuclides of Actinium -225.
  • the formulated 225 Ac-labeled precursor can be sterile filtered.
  • the radioactive labeled precursor obtained in step (7) has a radiochemical purity of more than about 95%, more preferably more than 97%.
  • the radiochemical purity can be 99% or more than 99%.
  • a sample from the obtained 225 Ac-labeled precursor solution or the formulated 225 Ac-labeled precursor solution is subjected to a bacterial endotoxin test, such as the Limulus amebocyte lysate (LAL) assay.
  • a bacterial endotoxin test such as the Limulus amebocyte lysate (LAL) assay.
  • microorganisms may affect the stability of drug substances due to their propensity to degrade/metabolize peptides.
  • Microbiological examinations involve the bioburden control (see European Pharmacopeia, Ph. Eur 2.6. 12) and content of bacterial endotoxins (see European Pharmacopeia, Ph Eur. 2.6.14).
  • the microbial enumeration tests for total aerobic microbial counts (TAMC) and total yeast and mold counts (TYMC) must adhere to the acceptance criteria of 103 CFU/g and 102 CFU/g for bulk material and 102 CFU/g and 101 CFU per container for chemical precursors packed in single and multi-dose containers, respectively.
  • Bacterial endotoxin can be determined by the gel-clot or photometric methods (turbidimetric and chromogenic techniques) and acceptance criteria are limited to a maximum 100 lU/g for bulk material or maximum 10 IU per container for chemical precursors packed in single-dose and multidose containers.
  • the method of the present invention further comprises the step
  • the quality control preferably comprises radio thin layer chromatography (radio TLC) and/or radio high-performance liquid chromatography (HPLC) analysis.
  • radio TLC radio thin layer chromatography
  • HPLC radio high-performance liquid chromatography
  • the present invention provides the 225 Ac-labeled precursor obtained in a method of the present invention for use in the treatment of cancer.
  • the treatment preferably comprises targeted alpha therapy (TAT) of cancer.
  • TAT targeted alpha therapy
  • Targeted alpha-particle therapy is a method of targeted radionuclide therapy of various cancers. It employs radioactive substances which undergo alpha decay to treat diseased tissue at close proximity. TAT has the potential to provide highly targeted treatment, especially to microscopic tumor cells.
  • the 225 Ac-labeled precursor is a prostate-specific membrane antigen (PSMA)-targeted ligand, preferably 225 Ac-labeled PSMA-617 or 225 Ac-labeled PSMA I&T, and the cancer is prostate cancer.
  • PSMA prostate-specific membrane antigen
  • the 225 Ac-labeled precursor is a somatostatin receptor-targeted ligand, preferably 225 Ac-labeled DOTA-octreotate ( 225 Ac-labeled DOTA-TATE) or 225 Ac-labeled DOTA-octreotide ( 225 Ac-labeled DOTA-TOC), and the cancer are neuroendocrine tumors. Kits for radioactive labeling
  • the present invention provides a kit for radioactive labeling of a precursor with 225 Ac.
  • Said kit comprises
  • the precursor is preferably a biomolecule comprising a chelator suitable to complex 225 Ac, which is preferably connected to the biomolecule.
  • suitable chelators are DOTA, DOTA-GA or MACROPA.
  • the precursor is preferably a prostate-specific membrane antigen (PSMA)-targeted ligand, such as PSMA I&T or PSMA-617.
  • PSMA prostate-specific membrane antigen
  • the precursor is preferably a somatostatin receptor- targeted ligand, such as DOTA-TOC or DOTA-TATE.
  • PSMA-617 and PSMA I&T are applied for targeted alpha-therapy (TAT) of prostate cancer.
  • TAT targeted alpha-therapy
  • the current state-of-the- art labeling procedure was previously published (Hooijman et al, 2021) and is also applied in a slightly modified form in magnetic resonance imaging (MRI) for patient care:
  • the precursor PSMA I&T
  • an organic solvent with a concentration of 10 nmol per MBq 225 Ac.
  • An aqueous solution of a TRIS (tris(hydroxymethyl)aminomethane, 0.1 M, pH 9) is added.
  • 225 Ac dissolved in 0.1 M aqueous HC1 is added.
  • the solution is heated for 5 min at 95°C in a microwave.
  • Quality control is performed by radio-thin layer chromatography (radio-TLC).
  • the product is sterile filtered and formulated in isotonic saline.
  • the precursor is dissolved in a suitable buffer and then freeze-dried.
  • the freeze-dried precursor is then reconstituted with water, whereupon 225 Ac is added. After heating for 5 min at 95°C in a micro-wave reactor, quality control is performed.
  • freeze-dried kits i.e. freeze-dried precursors
  • freeze-dried precursors can be stored over long time-periods
  • Kits have a long tradition in nuclear medicine. They also had a huge impact, facilitating the production of e.g. " m Tc-labeled compounds and lately 68 Ga-based radiopharmaceuticals (Lepareur, 2022).
  • the method disclosed herein can also be applied to other precursors, such as somatostatin receptor-targeted ligands, e.g. DOTA-TATE or DOTA-TOC.
  • somatostatin receptor-targeted ligands e.g. DOTA-TATE or DOTA-TOC.
  • Somatostatin analogues have been investigated for both diagnostic and treatment purposes in tumors that are known to express somatostatin receptors (SSTR).
  • SSTR somatostatin receptors
  • the macrocyclic peptide-based ligands DOTATATE ((DOTA°-Tyr 3 )octreotate) and DOTATOC ((DOTA 0 - Tyr 3 )octreotide) demonstrated excellent characteristics for 68 Ga-imaging and targeted therapy with lutetium-177, yttrium-90 and lately with actinium-225 in some types of neuroendocrine tumors.
  • Neuroendocrine tumors (NETs) are a class of slow-growing tumors that arise from cells distributed mainly in the lungs, gastrointestinal tract, or pancreas.
  • a unique feature of NETs is their overexpression of SSTR on the tumor cells, which has established the basis for both diagnostic imaging and peptide receptor radionuclide therapy.
  • the following examples and drawings illustrate the present invention without,
  • PSMA prostate-membrane antigen
  • Actinium-225 was provided by JRC Düsseldorf and then dissolved in 0.1 M HC1.
  • the precursor PSMA I&T was purchased by ITM Isotope Technologies Kunststoff SE (Munich Germany). All necessary solvents and other organic reagents were purchased from either, Alfa Aesar (Karlsruhe, Germany), Sigma-Aldrich (Munich, Germany) or VWR (Darmstadt, Germany).
  • High-performance liquid chromatography (HPLC) was performed on a Prominence HPLC system (Shimadzu, Kyoto, Japan) with a Photo Diode Array detector (Shimadzu) and a GABI Star detector (Raytest, Straub enhardt, Germany).
  • Activity quantification was performed using a 2480 WIZARD2 automatic gamma counter (PerkinElmer, Waltham, United States). Labeling experiments are performed in sealed and crimped glass vials (0.5-2.0 mL, Biotage Uppsala, Sweden), using a Biotage, Initiator + microwave reactor.
  • radiochemical purity is determined by radio-TLC (ITLC-SG paper, 0.5 M sodium citrate pH 5) in triplicate.
  • TLC-stripe is cut at a relate-to-front value (Rf-value) of 0.7.
  • the origin represents complexed 225 Ac whereas the front represents non-complexed 225 Ac.
  • the samples are analyzed after at least 30 minutes in a gamma-counter quantifying the gamma-energy of Francium -221. Additionally, one TLC-analysis is performed without cutting the stripe using a radio-TLC-scanner.
  • a 1 mM solution of PSMA-I&T is prepared using EtOH/water (1 : 1, v/v) or DMSO.
  • 500 pL of TRIS buffer (0. 1 M, pH 9) are mixed with 50 pL PSMA I&T (1 mM, 50 nmol), and 5.0 MBq of 225 Ac is added (10 pL, 0.1 M HC1).
  • the reaction vial is sealed and heated in a microwave reactor for 5 minutes. After cooling of the reaction vial, quality control is performed as described in Example 2.
  • pH is crucial for the efficient labeling reaction, and it can change during freeze-drying if volatile buffers are used, several kits were prepared and the pH was measured after reconstitution in 500 pL of water. Moreover, after reconstitution, 10 and 20 pL of 0.1 M HC1 was added, simulating the addition of 225 Ac.
  • TRIS buffer (0.1 M, pH 9.0) was used, as it represents the most- often used buffer for 225 Ac-labeling of PSMA-targeted ligands.
  • NaOAc buffer (0.1 M, pH 5.7) was described as a promising buffer for 225 Ac-labeling, which suits better to current GMP-guidelines (Pretze et al., 2021).
  • acetate/acetic acid is known from literature (see e.g. Baheti et al., 2010) to be removed in some parts during freeze-drying.
  • Kits were prepared, by lyophilisation of 30 nmol of PSMA I&T, together with (A) 500 pL NaOAc, 0.1 M, pH 3.7; (B) 500 pL NaOAc, 0.1 M, pH 4.7 and (C) 500 pL NaOAc, 0.1 M, pH 5.7, in three separate vials. After reconstitution in 500 pL of water, 225Ac (3.0 MBq, 3.0 pL, 0.1 M HC1) was added. The reaction vials were sealed and heated for 5 min at 95°C in a microwave reactor. After cooling quality control is performed as described above (see Example 2).
  • the radiochemical purity by radio-TLC was:
  • TRIS buffer was stable after freeze-drying, reconstitution and addition of 10 or 20 pL of 0.1 M HC1, as shown in Table 1.
  • the pH value of acetate buffer changed after freeze-drying, nevertheless labeling of the kits with 225 Ac was successful, see Table 1.
  • Kit labeling of PSMA I&T with 225 Ac was feasible using different buffers and pH values, as shown in Table 1.
  • Kits were prepared by lyophilisation of 30 nmol of DOTATOC (30 pL, 1 mM in water), together with aqueous buffers of:
  • the radiochemical purity by radio-TLC was:

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Dispersion Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a method for radioactive labeling of a precursor with 225Ac comprising freeze-drying the precursor before labeling. The present invention further relates to the 225Ac-labeled precursor obtained in said method for use in the treatment of cancer. The present invention also relates to a kit for radioactive labeling of a precursor with 225Ac.

Description

Labeling of biomolecules with Actinium-225 and medical uses thereof
The present invention relates to a method for radioactive labeling of a precursor with 225 Ac comprising freeze-drying the precursor before labeling. The present invention further relates to the 225 Ac-labeled precursor obtained in said method for use in the treatment of cancer. The present invention also relates to a kit for radioactive labeling of a precursor with 225 Ac.
BACKGROUND OF THE INVENTION
An important prerequisite in targeted radionuclide therapy of cancer is the delivery of cytotoxic doses of radiation to malignant cells. In principle, beta-emitting-, alpha-emitting- or Auger electron-emitting nuclides, tethered to a targeted biomolecule for therapeutic purposes can be used (Boyd et al., 2006). Beta radiation is a particulate radiation consisting of high-speed electrons, which lose their kinetic energy, while their track paths become increasingly contorted due to scattering. An exemplary 2 MeV beta particle has a rather long range of 1 cm in water. The linear energy transfer (LET), which represents the total amount of energy deposited per unit track length, is comparably low for beta particles with values around 0.2 keV/pm (Boyd et al., 2006).
Alpha particles are positively charged with a mass and charge equal to a helium nucleus. Their energies are higher compared to beta particles with values of 5-9 MeV, displaying a linear track length of 20 to 80 pm in water. Moreover, their higher LET (approx. 100 keV/pm) can result in more lethal double strand DNA breaks, compared to beta particles. In combination with the short range of alphas, the high energy discharge results in a very toxic effect to a relatively small field with very limited damage to surrounding normal tissue (Scheinberg et al., 2011).
In the past, the alpha emitter Actinium-225 (225Ac) was suggested as a radionuclide for targeted alpha therapy (TAT) of cancer (Morgenstern et al, 2018). Thereby 225 Ac is complexed by a suitable chelator, connected to a biological active molecule. After intravenous injection, the radiopharmaceutical binds to cancer cells and delivers the payload, resulting in irradiation of cancer cells, while minimizing damage to healthy tissues. 225 Ac (ti/2 = 9.9 d; 6 MeV alpha-particle) decay yields 4 alpha and 3 beta disintegrations (Robertson et al., 2018). The daughter nuclides with suitable gamma emissions, i.e. 221Fr and 213Bi, can be exploited for imaging or measuring drug distribution. The decay chain of 225 Ac is summarized below and depicted in Figure 1 :
221Fr (ti/2 = 4.8 min; 6 MeV a-particle; 218 keV y-emission)
217At (ti/2 = 32.3 ms; 7 MeV a -particle)
213Bi (ti/2 = 45.6 min; 6 MeV a -particle, 444 keV P' -particle, 440 keV y -emission)
213Po (ti/2 = 4.2 ps; 8 MeV a -particle),
2O9T1 (ti/2 = 2.2 m; 659 keV P'-particle),
209Pb (ti/2 = 3.25 h; 198 keV P' -particle)
209Bi (stable).
Despite the favorable decay properties of 225 Ac for TAT, one main challenge remains its low availability for research and clinical applications (Morgenstern et al., 2018). Currently, the primary source of 225 Ac is 229Th (ti/2 = 7340 years), originating mainly from radioactive waste of nuclear power plants. The production of 225 Actinium via this route, which still represents the most reliable source of international supply, mounts however only up to <100 GBq. This limited supply in combination with immense costs (300-600C per MBq), is still preventing 225 Ac to develop its full clinical potential (Morgenstern etal, 2018). Currently there are several research projects underway, examining alternative production strategies of 225 Ac, which mainly rely on high-energy accelerators, e.g. the irradiation of Ra-226 targets with protons, neutrons or gamma rays (Apostolidis et al., 2005).
For many years an additional main challenge was the development of suitable bifunctional chelators and standardized labeling procedures, suitable for clinical application (Thiele and Wilson, 2018). This can be explained by limited understanding of the 225 Ac coordination chemistry, which is further hampered by the fact that no non-radioactive isotopes of Actinium exist. This lack of knowledge made it difficult to develop ligands, forming stable complexes with 225 Ac and as a result, this task was and still is conducted by trial and error (Thiele and Wilson, 2018). Instability of 225 Ac-ligands in vivo results in accumulation of free 225 Ac in the liver and bone, causing potentially radiotoxic side effects (Scheinberg et al., 2011). To minimize damage to healthy tissues, 225 Ac needs to be bound kinetically inert to a chelator and labeling procedures with respective chelators need to be reliable and quantitative. In theory, free 225 Ac after incomplete labeling reactions can also be separated by a cartridge-based purification step, but due to the high costs and limited availability of this radionuclide this approach is very uneconomic.
Currently, one of the most often used chelator for complexation of 225 Ac, is the 12-membered macrocyclic ligand 1,4,7, 10-Tetraazacyclododecane-l,4,7,10-tetrayltetraacetic acid (DOTA) or the glutamic-acid substituted derivative (DOTA-GA) (Thiele and Wilson, 2018). There are numerous 225Ac-DOTA constructs which have been evaluated in preclinical studies and are also under evaluation in several clinical trials, e.g. for the treatment of leukemia (Jurcic etal., 2017), multiple myeloma, and prostate cancer (Sathekge et al., 2021). Despite these studies, DOTA- based chelators are not regarded as ideal chelators for complexation of 225 Ac, because the metal ion radius is too large for the cavity of this chelator (Thiele and Wilson, 2018). Even though, the in vivo stability is considered to be sufficiently high for in vivo applications, a main challenge of this size mismatch between 225 Ac and DOTA, results in difficulties during radiolabeling. In general, complexation of DOTA with 225 Ac requires harsh reaction conditions, including elevated temperatures, micro-wave assistance and long time periods (Thiele and Wilson, 2018). Noteworthy, much research effort is currently dedicated to the development of novel chelators, which allow complexation of 225 Ac under mild reaction conditions. This resulted for example in identification of the larger MACROPA chelator, allowing radiolabeling at room temperature within 5 minutes (Thiele et al., 2017).
There is a need in the art for improved means and methods for radio-labelling of active biomolecules with 225 Ac, which in particular allow their use as radionuclides for targeted alpha therapy (TAT) of cancer.
SUMMARY OF THE INVENTION
According to the present invention this object is solved by a method for radioactive labeling of a precursor with 225 Ac, said method comprising the steps of
(1) providing a precursor dissolved in a buffer solution;
(2) freeze-drying the precursor provided in (1) and thereby obtaining a powder of the freeze-dried precursor;
(3) reconstituting the freeze-dried precursor with water;
(4) providing 225 Ac in 0.04 M to 0.2 M HC1; (5) adding the 225 Ac of (4) to the reconstituted precursor of (3);
(6) heating to at least 75°C for a period of time; and
(7) obtaining the 225 Ac-labeled precursor
According to the present invention this object is solved by the 225 Ac-labeled precursor obtained in a method according to the present invention for use in the treatment of cancer.
According to the present invention this object is solved by a kit for radioactive labeling of a precursor with 225 Ac, said kit comprising
(i) a freeze-dried precursor obtained according to steps (1) and (2) of the method of the present invention, and
(ii) 225 Ac in 0.04 M to 0.2 M HC1,
(iii) optionally, water for reconstituting the freeze-dried precursor (i).
DESCRIPTION OF THE PREFERRED EMBODIMENTS OF THE INVENTION
Before the present invention is described in more detail below, it is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. For the purpose of the present invention, all references cited herein are incorporated by reference in their entireties.
Concentrations, amounts, and other numerical data may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. As an illustration, a numerical range of "0.5 to 10" should be interpreted to include not only the explicitly recited values of 0.5 to 10, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 0.5, 1, 1.5, 2, 2.5, 3, 4, 5 .... 8, 8.5, 9, 9.5, 10 and sub-ranges such as from 2 to 8, 4 to 5, etc. This same principle applies to ranges reciting only one numerical value, such as "less than 1 mm". Furthermore, such an interpretation should apply regardless of the breadth of the range or the characteristics being described.
Method for labeling of biomolecules with 225 Ac
As outlined above, the present invention provides a method for radioactive labeling of a precursor with 225 Ac.
Said method comprises the steps of:
(1) providing a precursor dissolved in a buffer solution;
(2) freeze-drying the precursor provided in (1) and thereby obtaining a powder of the freeze-dried precursor;
(3) reconstituting the freeze-dried precursor with water;
(4) providing 225 Ac in 0.04 M to 0.2 M HC1;;
(5) adding the 225 Ac of (4) to the reconstituted precursor of (3);
(6) heating to at least 75°C for a period of time; and
(7) obtaining the 225 Ac-labeled precursor.
Precursor
The precursor is preferably a biomolecule comprising a chelator suitable to complex 225 Ac.
The chelator is preferably connected to the biomolecule.
Suitable chelators are DOTA, DOTA-GA and MACROPA:
- DOTA:
1 ,4,7, 10-tetraazacyclododecane- 1 ,4,7, 10-tetrayltetraacetic acid
- DOTA-GA:
Glutamic-acid substituted derivative of DOTA.
- MACROPA:
6- [ [ 16-[(6-carboxypyridin-2-yl)m ethyl]- 1 ,4,10, 13 -tetraoxa-7, 16-diazacycl ooctadec-7- yl]methyl]-4-isothiocyanatopyridine-2-carboxylic acid (MACROPA).
PubChem CID: 135349208. In a preferred embodiment, the precursor is a prostate-specific membrane antigen (PSMA)- targeted ligand.
Preferred PSMA-targeted ligands are PSMA I&T, or PSMA-617.
- PSMA I&T:
Prostate-specific membrane antigen for imaging & therapy (PSMA I&T) is commercially available, such as from ITM Isotope Technologies Munich SE, Germany, or Scintomics Molecular, Applied Theranostics Technologies (SCI-att), Germany.
Synonym s : (R)-DOT AGA-(I-y )fk( Sub -KuE) .
Sequence: Suber- 1 -oyl-s-(DOTA-GA-3 -iodo-D-Tyr-D-Phe-D-Lys-OH)-8-oyl -s-(HO-Glu- ureido-Lys-OH).
Molecular Weight: 1497.5 (m.i.), 1498.4 (av.).
Molecular Formula: C63H92IN11O23.
Structure is shown in Figure 2 A.
- PSMA-617:
Vipivotide tetraxetan (PSMA-617) is a high potent prostate-specific membrane antigen (PSMA) inhibitor. PSMA-617 is commercially available, such as from Advanced Accelerator Applications (AAA, a Novartis company), France.
Structure is shown in Figure 2 A.
In a preferred embodiment, the precursor is a somatostatin receptor-targeted ligand.
Preferred somatostatin receptor-targeted ligands are octreotide or octreotate, more preferably DOTA-octreotide or DOTA-octreotate
Octreotide
Octreotide, sold under the brand name Sandostatin among others, is an octapeptide that mimics natural somatostatin pharmacologically. It binds predominantly to the somatostatin receptors SSTR2 and SSTR5.
CAS number: 83150-76-9
Sequence: H-D-Phe-Cys-Phe-D-Trp-Lys-Thr-Cys-Thr-ol
Molecular Weight: 1019.25 g mol-1.
Molecular Formula: C49H66N10O10S2. Structure is shown in Figure 2B.
- DOTA-octreotide (DOTA-TOC)
DOTA-TOC (DOTATOC, DOTA-octreotide, DOTA-(Tyr3)-octreotide, and DOTA(O)- Phe(l)-Tyr(3))octreotide) is a compound containing tyrosine3-octreotide, an SSR agonist, and the bifunctional chelator DOTA. Edotreotide is itse International Nonproprietary Name.
CAS number: 204318-14-9
Molecular Weight: 1421.64 g mol-1
Molecular Formula: C65H92N14O18S2.
Structure is shown in Figure 2B.
Octreotate
Octreotate or octreotide acid is a somatostatin analog that is closely related to octreotide. It is a somatostatin receptor peptide agonist.
Sequence: H-D-Phe-Cys-Phe-D-Trp-Lys-Thr-Cys-Thr-OH.
Molecular Weight: 1033.23 g mol-1.
Molecular Formula: C49H64N10O11S2.
Structure is shown in Figure 2C.
DOTA-octreotate (DOTA-TATE)
DOTA-TATE (DOTATATE, DOTA-octreotate, oxodotreotide, DOTA-(Tyr3)-octreotate, and DOTA-0-Tyr3-Octreotate) is a compound containing tyrosine3 -octreotate, an SSR agonist, and the bifunctional chelator DOTA (tetraxetan).
CAS number: 177943-89-4
Molecular Weight: 1435.63 g mol-1.
Molecular Formula: C65H90N14O19S2.
Structure is shown in Figure 2C.
In one embodiment, where the precursor is obtained from a commercial provider, it has been sterile filtered by the commercial provider. Furthermore, microbiological evaluations have been carried out by the commercial provider, such as bioburden control and/or the content of bacterial endotoxins has been determined.
Step (1)
In step 1, a precursor is provided. The precursor is dissolved in a buffer solution. In one embodiment, the buffer solution is a TRIS (tris(hydroxymethyl)aminomethane) buffer solution, preferably 0.1 M TRIS buffer having pH 7 to 9, more preferably 0.1 M TRIS pH 9.
In one embodiment, the buffer solution is an acetate buffer solution, preferably 0.1 M to IM acetate pH 4 to 6, more preferably 0.1 M acetate pH 5.7.
For example, preferably 0.1 M NaOAc (sodium acetate) buffer having pH 5.7.
In one embodiment, the buffer solution is an acetate buffer solution, preferably 0.1 M to IM acetate pH 3 to 6, more preferably 0.1 M acetate pH 3.7.
For example, preferably 0.1 M NaOAc (sodium acetate) buffer having pH 3.7.
In one embodiment, the buffer solution is a phosphate buffer solution, preferably 0. 1 M to 0.3 M phosphate pH 5 to 6, more preferably 0.2 M phosphate pH 5.7.
The buffer solutions can be sterile filtered before and/or after dissolving the precursor.
In a preferred embodiment, the buffer solution including the dissolved precursor is sterile filtered.
Step (2)
In step 2, the precursor is freeze-dried. A powder of the freeze-dried precursor is obtained.
In step 2, the precursor is freeze-dried while it is dissolved in buffer. The powder of the freeze-dried precursor also contains buffer salt(s).
In one embodiment, the freeze-drying is via lyophilization.
In one embodiment, the solution is sterile filtered before lyophilization.
Step (3)
In step 3, the freeze-dried precursor is reconstituted with water.
In one embodiment, the freeze-dried precursor is reconstituted with water soon after the freeze-drying, such as immediately. In one embodiment, there is a period of time between obtaining the powder of the freeze-dried precursor and reconstituting it for further use. This means the powder of the freeze-dried precursor can be stored before radiolabeling with 225 Ac.
The storage is preferably at about -20°C. The storage time depends on the shelf life of the precursor, which can be up to one year.
Said period of time can be in a range from hours to months, such as one to two months, but also up to one year.
Step (4)
In step 4, 225 Ac is provided.
225 Ac is provided in 0.04 M to 0.2 M HC1.
Preferably, 225 Ac is provided in 0.1 M HC1.
The activity or dosage of 225 Ac is usually in the range from 4 to 12 MBq.
Step (5)
In step 5, the 225 Ac of step (4) is added to the reconstituted precursor of (3).
In one embodiment, 8 MBq of 225 Ac is added.
The ratio of 225 Ac to reconstituted precursor is preferably 5 to 20 nmol per MBq 225 Ac, preferably 10 nmol per MBq 225 Ac.
Step (6)
In step 6, heating for a period of time is carried out.
Preferably, the heating is to at least about 75°C.
In a preferred embodiment, the heating in step (6) is carried out at about 95°C in a microwave reactor and the period of time is from about 5 to 10 min, preferably about 5 min.
In one embodiment, the heating in step (6) is carried out at about 75°C to about 85°C and the period of time is from about 20 to 30 min, preferably 75°C for about 20 min. Step (7)
In step 7, the 225 Ac-labeled precursor is obtained.
Step (7) preferably comprises cooling.
In one embodiment, the obtained 225 Ac-labeled precursor is formulated, preferably in isotonic saline, and/or sterile filtered.
The 225 Ac-labeled precursor obtained in step (7) can be formulated in isotonic saline. The formulation optionally contains one or more stabilizing agents and/or a co-chelator.
Examples for suitable stabilizing agents are:
- ascorbic acid, such as with a concentration of 10 to 100 mg per mL of the total formulation,
- gentisic acid (such as with a concentration of 1 to 5 mg per mL of the total formulation),
- methionine (such as with a concentration of 1 to 5 mg per mL of the total formulation)
An example for a suitable co-chelator is diethylenetriamine pentaacetate (DTP A), such as with a concentration of 0.01 to 0.03 mg per mL of the total formulation. Said co-chelator is capable of chelating radioactive daughter nuclides of Actinium -225.
In one embodiment, the formulated 225 Ac-labeled precursor can be sterile filtered.
Preferably, the radioactive labeled precursor obtained in step (7) has a radiochemical purity of more than about 95%, more preferably more than 97%. The radiochemical purity can be 99% or more than 99%.
In one embodiment, a sample from the obtained 225 Ac-labeled precursor solution or the formulated 225 Ac-labeled precursor solution is subjected to a bacterial endotoxin test, such as the Limulus amebocyte lysate (LAL) assay.
Microbiological examinations
The presence of microorganisms may affect the stability of drug substances due to their propensity to degrade/metabolize peptides. Microbiological examinations involve the bioburden control (see European Pharmacopeia, Ph. Eur 2.6. 12) and content of bacterial endotoxins (see European Pharmacopeia, Ph Eur. 2.6.14). The microbial enumeration tests for total aerobic microbial counts (TAMC) and total yeast and mold counts (TYMC) must adhere to the acceptance criteria of 103 CFU/g and 102 CFU/g for bulk material and 102 CFU/g and 101 CFU per container for chemical precursors packed in single and multi-dose containers, respectively. Bacterial endotoxin can be determined by the gel-clot or photometric methods (turbidimetric and chromogenic techniques) and acceptance criteria are limited to a maximum 100 lU/g for bulk material or maximum 10 IU per container for chemical precursors packed in single-dose and multidose containers.
In one embodiment, the method of the present invention further comprises the step
(8) quality control of the 225 Ac-labeled precursor.
The quality control preferably comprises radio thin layer chromatography (radio TLC) and/or radio high-performance liquid chromatography (HPLC) analysis.
Medical uses of the 225 Ac-labeled precursors
As outlined above, the present invention provides the 225 Ac-labeled precursor obtained in a method of the present invention for use in the treatment of cancer.
The treatment preferably comprises targeted alpha therapy (TAT) of cancer.
Targeted alpha-particle therapy (TAT) is a method of targeted radionuclide therapy of various cancers. It employs radioactive substances which undergo alpha decay to treat diseased tissue at close proximity. TAT has the potential to provide highly targeted treatment, especially to microscopic tumor cells.
In a preferred embodiment, the 225 Ac-labeled precursor is a prostate-specific membrane antigen (PSMA)-targeted ligand, preferably 225 Ac-labeled PSMA-617 or 225 Ac-labeled PSMA I&T, and the cancer is prostate cancer.
In one embodiment, the 225 Ac-labeled precursor is a somatostatin receptor-targeted ligand, preferably 225 Ac-labeled DOTA-octreotate (225 Ac-labeled DOTA-TATE) or 225 Ac-labeled DOTA-octreotide (225 Ac-labeled DOTA-TOC), and the cancer are neuroendocrine tumors. Kits for radioactive labeling
As outlined above, the present invention provides a kit for radioactive labeling of a precursor with 225 Ac.
Said kit comprises
(i) a freeze-dried precursor obtained according to steps (1) and (2) of the method of the present invention, and
(ii) 225 Ac in 0.04 M to 0.2 M HCL, preferably 0.1 M HC1,
(iii) optionally, water for reconstituting the freeze-dried precursor (i).
The precursor is preferably a biomolecule comprising a chelator suitable to complex 225 Ac, which is preferably connected to the biomolecule.
As discussed above, suitable chelators are DOTA, DOTA-GA or MACROPA.
As discussed above, the precursor is preferably a prostate-specific membrane antigen (PSMA)-targeted ligand, such as PSMA I&T or PSMA-617.
As discussed above, in one embodiment, the precursor is preferably a somatostatin receptor- targeted ligand, such as DOTA-TOC or DOTA-TATE.
Preferred embodiments
The 225 Ac-labeled prostate-membrane antigen (PSMA)-targeted ligands PSMA-617 and PSMA I&T are applied for targeted alpha-therapy (TAT) of prostate cancer. The current state-of-the- art labeling procedure was previously published (Hooijman et al, 2021) and is also applied in a slightly modified form in magnetic resonance imaging (MRI) for patient care:
In said prior method, the precursor (PSMA I&T) is dissolved in an organic solvent with a concentration of 10 nmol per MBq 225 Ac. An aqueous solution of a TRIS (tris(hydroxymethyl)aminomethane, 0.1 M, pH 9) is added. 225 Ac (dissolved in 0.1 M aqueous HC1) is added. The solution is heated for 5 min at 95°C in a microwave. Quality control is performed by radio-thin layer chromatography (radio-TLC). The product is sterile filtered and formulated in isotonic saline.
For a prior art method, see also Kratochwil et al. (2016). The inventors herein disclose a new and easy method to facilitate the radio-labeling procedure of PSMA I&T, as an example for other small biomolecules, with 225 Ac by employing freeze- dried kits, which can promote widespread of Ac-225 labeling procedure.
For the inventors’ protocol, the precursor is dissolved in a suitable buffer and then freeze-dried. The freeze-dried precursor is then reconstituted with water, whereupon 225 Ac is added. After heating for 5 min at 95°C in a micro-wave reactor, quality control is performed.
The method as disclosed herein has the following advantages:
1) Faster and minimized protocol. Only water and 225 Ac is required for reconstitution of the kit.
2) This bears potential advantages with respect to GMP-guidelines. Solutions can be sterile filtered before lyophilization, resulting in a low risk of contamination.
3) No sophisticated methods. The radiolabeling can also easily be performed by technicians.
4) The freeze-dried kits (i.e. freeze-dried precursors) can be stored over long time-periods
5) The radiolabeling can be performed on-site. Since the stability of 225 Ac-radiopharmaceuti cals is limited, this could hamper their widespread use (Hooijman et al., 2021). Therefore, there might be a huge demand for sites to produce their 225Ac-radiopharmaceuticals on-site in an easy and fast way.
6) Kits have a long tradition in nuclear medicine. They also had a huge impact, facilitating the production of e.g. "mTc-labeled compounds and lately 68Ga-based radiopharmaceuticals (Lepareur, 2022).
The method disclosed herein can also be applied to other precursors, such as somatostatin receptor-targeted ligands, e.g. DOTA-TATE or DOTA-TOC.
Somatostatin analogues have been investigated for both diagnostic and treatment purposes in tumors that are known to express somatostatin receptors (SSTR). In the past the macrocyclic peptide-based ligands DOTATATE ((DOTA°-Tyr3)octreotate) and DOTATOC ((DOTA0- Tyr3)octreotide) demonstrated excellent characteristics for 68Ga-imaging and targeted therapy with lutetium-177, yttrium-90 and lately with actinium-225 in some types of neuroendocrine tumors. Neuroendocrine tumors (NETs) are a class of slow-growing tumors that arise from cells distributed mainly in the lungs, gastrointestinal tract, or pancreas. A unique feature of NETs is their overexpression of SSTR on the tumor cells, which has established the basis for both diagnostic imaging and peptide receptor radionuclide therapy. The following examples and drawings illustrate the present invention without, however, limiting the same thereto.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Decay chain of 225 Ac.
The scheme was adapted from Thiele and Wilson (2018).
Figure 2. Biomolecules as precursors.
A) The prostate-membrane antigen (PSMA)-targeted ligands PSMA-617 and PSMA I&T radiolabeled with 225 Ac.
B) The somatostatin receptor-targeted ligand octreotide and DOTA-TOC which is radiolabeled with 225 Ac.
C) The somatostatin receptor-targeted ligand octreotate and DOTA-TATE which is radiolabeled with 225 Ac.
Figure 3. Quality control: Radio-TLC of225Ac-PSMA I&T after Kit-Labeling.
Figure 4. Exemplary radio-HPLC analysis of 225Ac-PSMA I&T.
Performed on a Shimadzu system, equipped with a variable wavelength detector (both Shimadzu) and a gamma-detector Gabi Star (Elysia-raytest, Straubenhardt, Germany). A) shows detection at the radiochannel and B) detection at 220 nm. Water/0. 1% TFA (solvent A) and MeCN/0.1% TFA (solvent B) served as mobile phases, a Nucleosil 100-5 C18 column of 125 x 4 mm was used as stationary phase. 15 pl of product solution were injected and the following linear solvent gradient was applied: 10-70% B in 15 min, 95 % B for 5 min (flow rate = 1 ml/min). Chemical impurities were monitored at 220 nm.
Figure 5. Quality control: Radio-TLC of225Ac-DOTATOC after Kit-Labeling.
Figure 6. Exemplary radio-HPLC analysis of 225Ac-DOTATOC.
Performed on a Shimadzu system, equipped with a variable wavelength detector (both Shimadzu) and a gamma-detector Gabi Star (Elysia-raytest, Straubenhardt, Germany). A) shows detection at the radiochannel and B) detection at 220 nm. EXAMPLES
EXAMPLE 1 Materials and Methods
Actinium-225 was provided by JRC Karlsruhe and then dissolved in 0.1 M HC1. The precursor PSMA I&T was purchased by ITM Isotope Technologies Munich SE (Munich Germany). All necessary solvents and other organic reagents were purchased from either, Alfa Aesar (Karlsruhe, Germany), Sigma-Aldrich (Munich, Germany) or VWR (Darmstadt, Germany). High-performance liquid chromatography (HPLC) was performed on a Prominence HPLC system (Shimadzu, Kyoto, Japan) with a Photo Diode Array detector (Shimadzu) and a GABI Star detector (Raytest, Straub enhardt, Germany). Eluents for all HPLC operations were water (solvent A) and acetonitrile (solvent B), both containing 0.1% trifluoroacetic acid. ANucleosil 100 C18 (125 x 4.6 mm, 5 pm particle size) column (CS Chromatographic Service, Langerwehe, Germany) was used for analytical measurements at a flow rate of 1 mL/min. Radio-Thin layer chromatography (radio-TLC) was performed using 0.5 M aqueous sodium citrate pH 5 and ITCL-SG paper (0.5 x 6 cm). TLC was then analyzed using a radio-TLC scanner from Bioscan (Eckert and Ziegler, Brussels, Belgium) and the Bio-Chrom Lite software. Activity quantification was performed using a 2480 WIZARD2 automatic gamma counter (PerkinElmer, Waltham, United States). Labeling experiments are performed in sealed and crimped glass vials (0.5-2.0 mL, Biotage Uppsala, Sweden), using a Biotage, Initiator+ microwave reactor.
EXAMPLE 2 Quality Control of 225 Ac-labeled radiopharmaceuticals
After the labeling reaction, radiochemical purity is determined by radio-TLC (ITLC-SG paper, 0.5 M sodium citrate pH 5) in triplicate. After analysis the TLC-stripe is cut at a relate-to-front value (Rf-value) of 0.7. The origin represents complexed 225 Ac whereas the front represents non-complexed 225 Ac. To ensure formation of an equilibrium between 225 Ac and daughter nuclides, the samples are analyzed after at least 30 minutes in a gamma-counter quantifying the gamma-energy of Francium -221. Additionally, one TLC-analysis is performed without cutting the stripe using a radio-TLC-scanner.
In order to determine potential degradation of the radiotracer, HPLC-analysis is performed using the following parameters: Water/0.1% TFA (solvent A) and MeCN/0.1% TFA (solvent B) served as mobile phases, a Nucleosil 100-5 C18 column of 125 x 4.6 mm was used as stationary phase. 5-50 pl of product solution were injected and the following linear solvent gradient was applied: 10-70 % B in 15 min, 95 % B for 5 min, and re-equilibration at 10% B for 5 min (flow rate = 1 ml/min). Chemical impurities were monitored at 220 and 254 nm Please note that analysis of 225 Ac-labeled radiopharmaceuticals results in an injection peak at the radioactivity channel. This injection peak is a result of the daughter nuclides and do not represent free 225 Ac.
EXAMPLE 3 State-of-the-Art labeling reaction
A 1 mM solution of PSMA-I&T is prepared using EtOH/water (1 : 1, v/v) or DMSO. In the reaction vial, 500 pL of TRIS buffer (0. 1 M, pH 9) are mixed with 50 pL PSMA I&T (1 mM, 50 nmol), and 5.0 MBq of 225 Ac is added (10 pL, 0.1 M HC1). The reaction vial is sealed and heated in a microwave reactor for 5 minutes. After cooling of the reaction vial, quality control is performed as described in Example 2. The radiochemical purity by radio-TLC was determined to be 98.7±0.3% (n=3). Radiochemical purity as determined by radio-HPLC was >97%.
EXAMPLE 4 Freeze drying experiments
As the pH is crucial for the efficient labeling reaction, and it can change during freeze-drying if volatile buffers are used, several kits were prepared and the pH was measured after reconstitution in 500 pL of water. Moreover, after reconstitution, 10 and 20 pL of 0.1 M HC1 was added, simulating the addition of 225 Ac.
In a first set of experiment TRIS buffer (0.1 M, pH 9.0) was used, as it represents the most- often used buffer for 225Ac-labeling of PSMA-targeted ligands. Recently, also NaOAc buffer (0.1 M, pH 5.7) was described as a promising buffer for 225Ac-labeling, which suits better to current GMP-guidelines (Pretze et al., 2021). However, acetate/acetic acid is known from literature (see e.g. Baheti et al., 2010) to be removed in some parts during freeze-drying.
Results are shown in Table 1.
Table 1
Figure imgf000017_0001
Figure imgf000018_0001
EXAMPLE 5
A kit was prepared, by lyophilisation of 500 pL of TRIS buffer (0.1 M, pH 9) and 50 nmol of PSMA I&T. After reconstitution in 500 pL of water, 225 Ac (5.0 MBq, 20 pL, 0.1 M HC1) was added. The reaction vial was sealed and heated for 5 min at 95°C in a microwave reactor. After cooling of the reaction vial, quality control was performed as described above (see Example 2). The radiochemical purity by radio-TLC was determined to be 99.3±0.1% (n=3). Radiochemical purity as determined by radio-HPLC was >97%.
EXAMPLE 6
Kits were prepared, by lyophilisation of 30 nmol of PSMA I&T, together with (A) 500 pL NaOAc, 0.1 M, pH 3.7; (B) 500 pL NaOAc, 0.1 M, pH 4.7 and (C) 500 pL NaOAc, 0.1 M, pH 5.7, in three separate vials. After reconstitution in 500 pL of water, 225Ac (3.0 MBq, 3.0 pL, 0.1 M HC1) was added. The reaction vials were sealed and heated for 5 min at 95°C in a microwave reactor. After cooling quality control is performed as described above (see Example 2).
The radiochemical purity by radio-TLC was:
(A) 99.5±0.1% (n=3),
(B) 99.8±0.0% (n=3),
(C) 99.6±0.1% (n=3).
Short summary of Examples 4 to 6:
The pH value of TRIS buffer was stable after freeze-drying, reconstitution and addition of 10 or 20 pL of 0.1 M HC1, as shown in Table 1. The pH value of acetate buffer changed after freeze-drying, nevertheless labeling of the kits with 225 Ac was successful, see Table 1.
Kit labeling of PSMA I&T with 225 Ac was feasible using different buffers and pH values, as shown in Table 1.
An excellent radiochemical purity of >99% was reached in all experiment for different kitpreparations (the cut-off for patient-application is 97%). The radiochemical purity was similar to the state-of-the-art procedure, as described in Example 3.
The identity of 225Ac-PSMA I&T could be confirmed by radio-HPLC, after kit-labeling.
EXAMPLE ?
We developed a kit-based approach for labeling the somatostatin receptor-targeted ligands DOTATOC and DOTATATE with actinium-225 for application in targeted alpha therapy of neuroendocrine tumors. The technical equipment, reagents and methods are identical to the ones described above for actinium-labeling of PSMA-ligands, see Examples 1-6.
Kits were prepared by lyophilisation of 30 nmol of DOTATOC (30 pL, 1 mM in water), together with aqueous buffers of:
(A) 500 pL NaOAc (1.0 M, pH 3.7),
(B) 500 pL TRIS (0.1 M, pH 9.0),
(C) 500 pL phosphate buffer (0.2 M, pH 5.7).
The radiochemical purity by radio-TLC was:
(A) 99.7±0.0% (n=3)
(B) 80.7±1.0% (n=3)
(C) 81.0±0.2% (n=3)
In radio-HPLC analysis, no degradation of the ligands was determined after labeling, see Figure 6. The features disclosed in the foregoing description, in the claims and/or in the accompanying drawings may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof.
REFERENCES
Apostolidis C, Molinet R, McGinley J, Abbas K, Mollenbeck J, Morgenstern A. Cyclotron production of Ac-225 for targeted alpha therapy. Appl Radiat Isot. 2005;62(3):383-7.
Baheti A, Kumar L, Bansal A K. Excipients used in lyophilization of small molecules. Journal of Excipients and Food Chemicals, [S.I.], v. 1, n. 1, p. 41-54, June 2010. ISSN 21502668.
Boyd M, Ross SC, Dorrens J, Fullerton NE, Tan KW, Zalutsky MR, et al. Radiation-induced biologic bystander effect elicited in vitro by targeted radiopharmaceuticals labeled with alpha- , beta-, and auger electron-emitting radionuclides. J Nucl Med. 2006;47(6): 1007-15.
Hooijman EL, Chalashkan Y, Ling SW, Kahyargil FF, Segbers M, Bruchertseifer F, et al. Development of [(225)Ac]Ac-PSMA-I&T for Targeted Alpha Therapy According to GMP Guidelines for Treatment of mCRPC. Pharmaceutics. 2021 ; 13(5).
Jurcic J, Levy M, Park J, Ravandi F, Perl A, Pagel J, et al. Phase I trial of alpha-particle immunotherapy with 225Ac-lintuzumab and low-dose cytarabine in patients age 60 or older with untreated acute myeloid leukemia. Journal of Nuclear Medicine. 2017;58(supplement 1):456-.
Kratochwil C, Bruchertseifer F, Giesel FL, Weis M, Verburg FA, Mottaghy F, Kopka K, Apostolidis C, Haberkron U, Morgenstern A. 225Ac-PSMA-617 for PSMA targeting alpharadiation therapy of patients with metastatic castration-resistant prostate cancer. J Nucl Med., 2016, 1-20.
Lepareur N. Cold Kit Labeling: The Future of (68)Ga Radiopharmaceuticals? Front Med (Lausanne). 2022;9:812050.
Morgenstern A, Apostolidis C, Kratochwil C, Sathekge M, Krolicki L, Bruchertseifer F. An Overview of Targeted Alpha Therapy with (225)Actinium and (213)Bismuth. Curr Radiopharm. 2018;l l(3):200-8. Pretze M, Kunkel F, Runge R, Freudenberg R, Braune A, Hartmann H, et al. Ac-EAZY! Towards GMP-Compliant Module Syntheses of (225)Ac-Labeled Peptides for Clinical Application. Pharmaceuticals (Basel). 2021;14(7).
Robertson AKH, Ramogida CF, Schaffer P, Radchenko V. Development of (225)Ac Radiopharmaceuticals: TRIUMF Perspectives and Experiences. Curr Radi opharm. 2018;l l(3):156-72.
Sathekge MM, Bruchertseifer F, Vorster M, Morgenstern A, Lawai IO. Global experience with PSMA-based alpha therapy in prostate cancer. Eur J Nucl Med Mol Imaging. 2021;49(l):30- 46.
Scheinberg DA, McDevitt MR. Actinium-225 in targeted alpha-particle therapeutic applications. Curr Radi opharm. 2011;4(4):306-20.
Thiele NA, Brown V, Kelly JM, Amor-Coarasa A, Jermilova U, MacMillan SN, et al. An Eighteen-Membered Macrocyclic Ligand for Actinium-225 Targeted Alpha Therapy. Angew Chem Int Ed Engl. 2017;56(46): 14712-7.
Thiele NA, Wilson JJ. Actinium-225 for Targeted a Therapy: Coordination Chemistry and Current Chelation Approaches. Cancer Biother Radiopharm. 2018;33(8):336-48.
Zielinska B, Apostolidis C, Bruchertseifer F, Morgenstern A (2007): An Improved Method for the Production of Ac-225/Bi-213 from Th-229 for Targeted Alpha Therapy, Solvent Extraction and Ion Exchange, 25:3, 339-349.

Claims

Claims
1. A method for radioactive labeling of a precursor with 225 Ac, said method comprising the steps of:
(1) providing a precursor dissolved in a buffer solution;
(2) freeze-drying the precursor provided in (1) and thereby obtaining a powder of the freeze-dried precursor;
(3) reconstituting the freeze-dried precursor with water;
(4) providing 225 Ac in 0.04 M to 0.2 M HC1;
(5) adding the 225 Ac of (4) to the reconstituted precursor of (3);
(6) heating to at least 75°C for a period of time; and
(7) obtaining the 225 Ac-labeled precursor.
2. The method of claim 1, wherein the precursor is a biomolecule comprising a chelator suitable to complex 225 Ac, which is preferably connected to the biomolecule, such as l,4,7,10-tetraazacyclododecane-l,4,7,10-tetrayltetraacetic acid (DOTA), a glutamic- acid substituted derivative of DOTA (DOTA-GA), or 6-[[16-[(6-carboxypyridin-2- yl)methyl]-l,4, 10, 13-tetraoxa-7, 16-diazacyclooctadec-7-yl]methyl]-4-isothiocyanatopyridine- 2-carboxylic acid (MACROP A).
3. The method of claim 1 or 2, wherein the precursor is a prostate-specific membrane antigen (PSMA)-targeted ligand, such as PSMA I&T, or PSMA-617, or wherein the precursor is a somatostatin receptor-targeted ligand, such as DOTA-octreotide (DOTA-TOC), or DOTA-octreotate (DOTA-TATE).
4. The method of any one of claims 1 to 3, wherein the buffer solution in step (1) is a TRIS buffer solution, preferably 0.1 M TRIS pH 7 to 9, more preferably 0.1 M TRIS pH 9, an acetate buffer solution, preferably 0.1 M to 1 M acetate pH 3 to 6, more preferably 0.1 M acetate pH
5.7 or 1.0 M acetate pH 3.7, or a phosphate buffer solution, preferably 0.1 M to 0.3 M phosphate pH 5 to 6, more preferably 0.2 M phosphate pH 5.7.
5. The method of any one of claims 1 to 4, wherein the ratio of 225 Ac to reconstituted precursor is 5-20 nmol per MBq 225 Ac, preferably 10 nmol per MBq 225 Ac, and/or wherein 225 Ac is provided in 0.1 M HC1.
6. The method of any one of claims 1 to 5, wherein the heating in step (6) is carried out at about 95°C in a microwave reactor and the period of time is from about 5 to 10 min, preferably about 5 min, and/or wherein the heating in step (6) is carried out at about 75°C to about 85°C and the period of time is from about 20 to 30 min, preferably 75°C for about 20 min.
7. The method of any one of claims 1 to 6, wherein the radioactive labeled precursor obtained in step (7) has a radiochemical purity of more than about 95%, preferably more than 97%, and/or wherein the 225 Ac-labeled precursor obtained in step (7) is formulated, preferably in isotonic saline.
8. The method of any one of claims 1 to 7, further comprising the step
(8) quality control of the 225 Ac-labeled precursor, wherein, preferably, the quality control comprises radio thin layer chromatography (radio TLC) and/or high-performance liquid chromatography (HPLC) analysis.
9. The method of any one of the preceding claims, wherein between obtaining the powder of the freeze-dried precursor in (2) and reconstituting it in step (3) is a period of time, such as in a range from hours to months.
10. The method of any one of the preceding claims, comprising sterile filtering of the solutions, such as of the buffer solution of (1), the precursor dissolved in the buffer solution in step (1), and/or the precursor of (1), and/or comprising microbiological examinations, including bioburden control and/or content of bacterial endotoxins, such as of the precursor of (1) or the 225 Ac-labeled precursor obtained in step (7).
11. The 225 Ac-labeled precursor obtained in a method of any one of claims 1 to 10 for use in the treatment of cancer.
12. The 225 Ac-labeled precursor for use according to claim 11, wherein the treatment comprises targeted alpha therapy (TAT) of cancer.
13. The 225 Ac-labeled precursor for use according to claim 11 or 12, wherein the 225 Ac- labeled precursor is a prostate-specific membrane antigen (PSMA)-targeted ligand, preferably 225 Ac-labeled PSMA-617 or 225 Ac-labeled PSMA I&T, and wherein the cancer is prostate cancer, or wherein the 225 Ac-labeled precursor is a somatostatin receptor-targeted ligand, preferably 225 Ac-labeled DOTA-octreotate (DOTA-TATE) or 225 Ac-labeled DOTA-octreotide (DOTA- TOC), and wherein the cancer are neuroendocrine tumors.
14. A kit for radioactive labeling of a precursor with 225 Ac, said kit comprising
(i) a freeze-dried precursor obtained according to steps (1) and (2) of the method of any one of claims 1 to 10, and
(ii) 225 Ac in 0.04 M to 0.2 M HCL, preferably 0.1 M HC1,
(iii) optionally, water for reconstituting the freeze-dried precursor (i).
15. The kit of claim 14, wherein the precursor is a biomolecule comprising a chelator suitable to complex 225 Ac, which is preferably connected to the biomolecule, such as l,4,7,10-tetraazacyclododecane-l,4,7,10-tetrayltetraacetic acid (DOTA), a glutamic- acid substituted derivative of DOTA (DOTA-GA), or 6-[[16-[(6-carboxypyridin-2- yl)methyl]-l,4, 10, 13-tetraoxa-7, 16-diazacyclooctadec-7-yl]methyl]-4-isothiocyanatopyridine- 2-carboxylic acid (MACROP A), and/or wherein the precursor is preferably a prostate-specific membrane antigen (PSMA)- targeted ligand, such as PSMA I&T, or PSMA-617, or a somatostatin receptor-targeted ligand, such as DOTA-TOC or DOTA-TATE.
PCT/EP2023/082190 2022-11-23 2023-11-17 Labeling of biomolecules with actinium-225 and medical uses thereof WO2024110329A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22209040 2022-11-23
EP22209040.9 2022-11-23

Publications (1)

Publication Number Publication Date
WO2024110329A1 true WO2024110329A1 (en) 2024-05-30

Family

ID=84361528

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/082190 WO2024110329A1 (en) 2022-11-23 2023-11-17 Labeling of biomolecules with actinium-225 and medical uses thereof

Country Status (1)

Country Link
WO (1) WO2024110329A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023037337A1 (en) * 2021-09-13 2023-03-16 Jubilant Radiopharma Pharmaceutical compositions for imaging, diagnosis and treatment of cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023037337A1 (en) * 2021-09-13 2023-03-16 Jubilant Radiopharma Pharmaceutical compositions for imaging, diagnosis and treatment of cancer

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
APOSTOLIDIS CMOLINET R, MCGINLEY JABBAS KMOLLENBECK JMORGENSTERN A: "Cyclotron production of Ac-225 for targeted alpha therapy", APPL RADIAT ISOT., vol. 62, no. 3, 2005, pages 383 - 7, XP004686574, DOI: 10.1016/j.apradiso.2004.06.013
BAHETI AKUMAR LBANSAL A K: "Excipients used in lyophilization of small molecules", JOURNAL OF EXCIPIENTS AND FOOD CHEMICALS, [S.L., vol. 1, no. 1, June 2010 (2010-06-01), pages 41 - 54, ISSN: 21502668
BOYD MROSS SCDORRENS JFULLERTON NETAN KWZALUTSKY MR ET AL.: "Radiation-induced biologic bystander effect elicited in vitro by targeted radiopharmaceuticals labeled with alpha-, beta-, and auger electron-emitting radionuclides", J NUCL MED., vol. 47, no. 6, 2006, pages 1007 - 15
CAS , no. 177943-89-4
HOOIJMAN ELCHALASHKAN YLING SWKAHYARGIL FFSEGBERS MBRUCHERTSEIFER F ET AL.: "Development of [(225)Ac]Ac-PSMA-I&T for Targeted Alpha Therapy According to GMP Guidelines for Treatment of mCRPC", PHARMACEUTICS, vol. 13, no. 5, 2021
HOOIJMAN ELINE L. ET AL: "Development of [225Ac]Ac-PSMA-I&T for Targeted Alpha Therapy According to GMP Guidelines for Treatment of mCRPC", PHARMACEUTICS, vol. 13, no. 5, 13 May 2021 (2021-05-13), CH, pages 715, XP093043803, ISSN: 1999-4923, DOI: 10.3390/pharmaceutics13050715 *
JURCIC JLEVY MPARK JRAVANDI FPERL APAGEL J ET AL.: "Phase I trial of alpha-particle immunotherapy with Ac-lintuzumab and low-dose cytarabine in patients age 60 or older with untreated acute myeloid leukemia", JOURNAL OF NUCLEAR MEDICINE, vol. 58, 2017, pages 456
KRATOCHWIL CBRUCHERTSEIFER FGIESEL FLWEIS MVERBURG FAMOTTAGHY FKOPKA KAPOSTOLIDIS CHABERKRON UMORGENSTERN A: "225Ac-PSMA-617 for PSMA targeting alpha-radiation therapy of patients with metastatic castration-resistant prostate cancer", J NUCL MED., 2016, pages 1 - 20
LEPAREUR N.: "Cold Kit Labeling: The Future of (68)Ga Radiopharmaceuticals?", FRONT MED (LAUSANNE, vol. 9, 2022, pages 812050
MORGENSTERN AAPOSTOLIDIS CKRATOCHWIL CSATHEKGE MKROLICKI LBRUCHERTSEIFER F: "An Overview of Targeted Alpha Therapy with (225)Actinium and (213)Bismuth", CURR RADIOPHARM., vol. 11, no. 3, 2018, pages 200 - 8
MUKHERJEE ARCHANA: "An Update on Extemporaneous Preparation of Radiopharmaceuticals Using Freeze-Dried Cold Kits", MINI-REVIEWS IN MEDICINAL CHEMISTRY, vol. 21, no. 11, 14 December 2020 (2020-12-14), pages 1322 - 1336, XP009544176, ISSN: 1389-5575, DOI: 10.2174/1389557520999201214233634 *
PRETZE MKUNKEL FRUNGE RFREUDENBERG RBRAUNE AHARTMANN H ET AL.: "Ac-EAZY! Towards GMP-Compliant Module Syntheses of (225)Ac-Labeled Peptides for Clinical Application", PHARMACEUTICALS (BASEL, vol. 14, no. 7, 2021
ROBERTSON AKHRAMOGIDA CFSCHAFFER PRADCHENKO V: "Development of (225)Ac Radiopharmaceuticals: TRILTMF Perspectives and Experiences", CURR RADIOPHARM., vol. 11, no. 3, 2018, pages 156 - 72
SATHEKGE MMBRUCHERTSEIFER FVORSTER MMORGENSTERN ALAWAL IO: "Global experience with PSMA-based alpha therapy in prostate cancer", EUR J NUCL MED MOL IMAGING., vol. 49, no. 1, 2021, pages 30 - 46, XP037651326, DOI: 10.1007/s00259-021-05434-9
SCHEINBERG DAMCDEVITT MR: "Actinium-225 in targeted alpha-particle therapeutic applications", CURR RADIOPHARM., vol. 4, no. 4, 2011, pages 306 - 20, XP093044097, DOI: 10.2174/1874471011104040306
THIELE NABROWN VKELLY JMAMOR-COARASA AJERMILOVA UMACMILLAN SN ET AL.: "An Eighteen-Membered Macrocyclic Ligand for Actinium-225 Targeted Alpha Therapy", ANGEW CHEM INT ED ENGL., vol. 56, no. 46, 2017, pages 14712 - 7, XP055543785, DOI: 10.1002/anie.201709532
THIELE NAWILSON JJ: "Actinium-225 for Targeted a Therapy: Coordination Chemistry and Current Chelation Approaches", CANCER BIOTHER RADIOPHARM., vol. 33, no. 8, 2018, pages 336 - 48, XP055736216, DOI: 10.1089/cbr.2018.2494

Similar Documents

Publication Publication Date Title
Breeman et al. Radiolabelling DOTA-peptides with 68 Ga
Breeman et al. 68Ga-labeled DOTA-peptides and 68Ga-labeled radiopharmaceuticals for positron emission tomography: current status of research, clinical applications, and future perspectives
Tshibangu et al. Automated GMP compliant production of [18 F] AlF-NOTA-octreotide
Norenberg et al. 213Bi-[DOTA0, Tyr3] octreotide peptide receptor radionuclide therapy of pancreatic tumors in a preclinical animal model
EP1763372B1 (en) Radionuclides for medical use
Huclier-Markai et al. Optimization of reaction conditions for the radiolabeling of DOTA and DOTA-peptide with 44m/44Sc and experimental evidence of the feasibility of an in vivo PET generator
Wilbur et al. Preparation and evaluation of para-[211At] astatobenzoyl labeled anti-renal cell carcinoma antibody A6H F (ab′) 2. In vivo distribution comparison with para-[125I] iodobenzoyl labeled A6H F (ab′) 2
Mukherjee et al. Development of single vial kits for preparation of 68 Ga-labelled peptides for PET imaging of neuroendocrine tumours
Altai et al. On the prevention of kidney uptake of radiolabeled DARPins
RU2741794C2 (en) Radiopharmaceutical solutions with preferred properties
Stenberg et al. In situ generated 212Pb-PSMA ligand in a 224Ra-solution for dual targeting of prostate cancer sclerotic stroma and PSMA-positive cells
US7794691B2 (en) Radionuclides for medical use
Tolmachev et al. 114mIn, a candidate for radionuclide therapy: low-energy cyclotron production and labeling of DTPA-D-phe-octreotide
Leyva Montana et al. Yttrium-90–current status, expected availability and applications of a high beta energy emitter
Saha Characteristics of specific radiopharmaceuticals
Khalid et al. Evaluation of carrier added and no carrier added 90 Y-EDTMP as bone seeking therapeutic radiopharmaceutical.
Li et al. Aminocarboxylate complexes and octreotide complexes with no carrier added 177Lu, 166Ho and 149Pm
WO2024110329A1 (en) Labeling of biomolecules with actinium-225 and medical uses thereof
Vimalnath et al. Ce‐141‐labeled DOTMP: A theranostic option in management of pain due to skeletal metastases
KR20180099705A (en) Isotope manufacturing method
Käkelä et al. Adventures in radiosynthesis of clinical grade [68 Ga] Ga-DOTA-Siglec-9
Couto et al. In vivo comparative study of hydroxyapatite labeled with different radioisotopes: evaluation of the scintigraphic images
Ghai et al. Optimizing the radiosynthesis of [68Ga] DOTA-MLN6907 peptide containing three disulfide cyclization bonds–a GCC specific chelate for clinical radiopharmaceuticals
Van Laere et al. Terbium radionuclides for theranostic applications in nuclear medicine: from atom to bedside
Beyer et al. Targeted cancer therapy: The potential role of terbium-149

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: 23806321

Country of ref document: EP

Kind code of ref document: A1