1
|
Van Laere C, Koole M, Deroose CM, de Voorde MV, Baete K, Cocolios TE, Duchemin C, Ooms M, Cleeren F. Terbium radionuclides for theranostic applications in nuclear medicine: from atom to bedside. Theranostics 2024; 14:1720-1743. [PMID: 38389843 PMCID: PMC10879862 DOI: 10.7150/thno.92775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Terbium features four clinically interesting radionuclides for application in nuclear medicine: terbium-149, terbium-152, terbium-155, and terbium-161. Their identical chemical properties enable the synthesis of radiopharmaceuticals with the same pharmacokinetic character, while their distinctive decay characteristics make them valuable for both imaging and therapeutic applications. In particular, terbium-152 and terbium-155 are useful candidates for positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging, respectively; whereas terbium-149 and terbium-161 find application in α- and β--/Auger electron therapy, respectively. This unique characteristic makes the terbium family ideal for the "matched-pair" principle of theranostics. In this review, the advantages and challenges of terbium-based radiopharmaceuticals are discussed, covering the entire chain from radionuclide production to bedside administration. It elaborates on the fundamental properties of terbium, the production routes of the four interesting radionuclides and gives an overview of the available bifunctional chelators. Finally, we discuss the preclinical and clinical studies as well as the prospects of this promising development in nuclear medicine.
Collapse
Affiliation(s)
- Camille Van Laere
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christophe M. Deroose
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Michiel Van de Voorde
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
| | - Kristof Baete
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Thomas E. Cocolios
- KU Leuven, Institute for Nuclear and Radiation Physics, Department of Physics and Astronomy, Leuven, Belgium
| | | | - Maarten Ooms
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Zubenko AD, Shchukina AA, Chernikova EY, Egorova BV, Ikonnikova IS, Priselkova AB, Larenkov AA, Bubenshchikov VB, Mitrofanov AA, Fedorov YV, Fedorova OA. Synthesis of new acyclic chelators H4aPyta and H6aPyha and their complexes with Cu 2+, Ga 3+, Y 3+, and Bi 3. Dalton Trans 2024; 53:1141-1155. [PMID: 38105658 DOI: 10.1039/d3dt03451h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
In this article, we present the synthesis and characterization of new acyclic pyridine-containing polyaminocarboxylate ligands H4aPyta and H6aPyha, which differ in structural rigidity and the number of chelating groups. Their abilities to form complexes with Cu2+, Ga3+, Y3+, and Bi3+ cations, as well as the stability of the complexes, were evaluated by potentiometric titration method, radiolabeling with the corresponding radionuclides, in vitro studies, mass spectrometry, and HPLC. The structures of the resulting complexes were determined using NMR spectroscopy and DFT calculations. The results obtained made it possible to evaluate the influence of the structural features of the complexes on their stability. The developed chelators H4aPyta and H6aPyha were proved to be promising for further research in the field of radiopharmaceuticals.
Collapse
Affiliation(s)
- Anastasia D Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Bayirta V Egorova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Irina S Ikonnikova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Anna B Priselkova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Anton A Larenkov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., Bld. 46, 123098 Moscow, Russian Federation
| | - Viktor B Bubenshchikov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., Bld. 46, 123098 Moscow, Russian Federation
| | - Artem A Mitrofanov
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| |
Collapse
|
3
|
Ramogida C, Price E. Transition and Post-Transition Radiometals for PET Imaging and Radiotherapy. Methods Mol Biol 2024; 2729:65-101. [PMID: 38006492 DOI: 10.1007/978-1-0716-3499-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Radiometals are an exciting class of radionuclides because of the large number of metallic elements available that have medically useful isotopes. To properly harness radiometals, they must be securely bound by chelators, which must be carefully matched to the radiometal ion to maximize radiolabeling performance and the stability of the resulting complex. This chapter focuses on practical aspects of radiometallation chemistry including chelator selection, radiolabeling procedures and conditions, radiolysis prevention, purification, quality control, requisite equipment and reagents, and useful tips.
Collapse
Affiliation(s)
- Caterina Ramogida
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada.
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada.
| | - Eric Price
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
4
|
Winter G, Hamp-Goldstein C, Fischer G, Kletting P, Glatting G, Solbach C, Herrmann H, Sala E, Feuring M, Döhner H, Beer AJ, Bunjes D, Prasad V. Optimization of Radiolabeling of a [ 90Y]Y-Anti-CD66-Antibody for Radioimmunotherapy before Allogeneic Hematopoietic Cell Transplantation. Cancers (Basel) 2023; 15:3660. [PMID: 37509321 PMCID: PMC10377894 DOI: 10.3390/cancers15143660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
For patients with acute myeloid leukemia, myelodysplastic syndrome, or acute lymphoblastic leukemia, allogeneic hematopoietic cell transplantation (HCT) is a potentially curative treatment. In addition to standard conditioning regimens for HCT, high-dose radioimmunotherapy (RIT) offers the unique opportunity to selectively deliver a high dose of radiation to the bone marrow while limiting side effects. Modification of a CD66b-specific monoclonal antibody (mAb) with a DTPA-based chelating agent should improve the absorbed dose distribution during therapy. The stability and radioimmunoreactive fraction of the radiolabeled mAbs were determined. Before RIT, all patients underwent dosimetry to determine absorbed doses to bone marrow, kidneys, liver, and spleen. Scans were performed twenty-four hours after therapy for quality control. A radiochemical purity of >95% and acceptable radioimmunoreactivity was achieved. Absorbed organ doses for the liver and kidney were consequently improved compared to reported historical data. All patients tolerated RIT well with no treatment-related acute adverse events. Complete remission could be observed in 4/5 of the patients 3 months after RIT. Two patients developed delayed liver failure unrelated to the radioimmunotherapy. The improved conjugation and radiolabeling procedure resulted in excellent stability, radiochemical purity, and CD66-specific radioimmunoreactivity of 90Y-labeled anti-CD66 mAb. RIT followed by conditioning and HCT was well tolerated. Based on these promising initial data, further prospective studies of [90Y]Y-DTPA-Bn-CHX-A″-anti-CD66-mAb-assisted conditioning in HCT are warranted.
Collapse
Affiliation(s)
- Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Gabriel Fischer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Hendrik Herrmann
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Elisa Sala
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Donald Bunjes
- Department of Internal Medicine III, Ulm University Medical Center, 89081 Ulm, Germany
| | - Vikas Prasad
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
- Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
5
|
Holik HA, Ibrahim FM, Elaine AA, Putra BD, Achmad A, Kartamihardja AHS. The Chemical Scaffold of Theranostic Radiopharmaceuticals: Radionuclide, Bifunctional Chelator, and Pharmacokinetics Modifying Linker. Molecules 2022; 27:3062. [PMID: 35630536 PMCID: PMC9143622 DOI: 10.3390/molecules27103062] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Therapeutic radiopharmaceuticals have been researched extensively in the last decade as a result of the growing research interest in personalized medicine to improve diagnostic accuracy and intensify intensive therapy while limiting side effects. Radiometal-based drugs are of substantial interest because of their greater versatility for clinical translation compared to non-metal radionuclides. This paper comprehensively discusses various components commonly used as chemical scaffolds to build radiopharmaceutical agents, i.e., radionuclides, pharmacokinetic-modifying linkers, and chelators, whose characteristics are explained and can be used as a guide for the researcher.
Collapse
Affiliation(s)
- Holis Abdul Holik
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Faisal Maulana Ibrahim
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Angela Alysia Elaine
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Bernap Dwi Putra
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Arifudin Achmad
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
- Oncology and Stem Cell Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
| | - Achmad Hussein Sundawa Kartamihardja
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
| |
Collapse
|
6
|
Lucio-Martínez F, Garda Z, Váradi B, Kálmán FK, Esteban-Gómez D, Tóth É, Tircsó G, Platas-Iglesias C. Rigidified Derivative of the Non-macrocyclic Ligand H 4OCTAPA for Stable Lanthanide(III) Complexation. Inorg Chem 2022; 61:5157-5171. [PMID: 35275621 PMCID: PMC8965877 DOI: 10.1021/acs.inorgchem.2c00501] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
The stability constants
of lanthanide complexes with the potentially
octadentate ligand CHXOCTAPA4–,
which contains a rigid 1,2-diaminocyclohexane scaffold functionalized
with two acetate and two picolinate pendant arms, reveal the formation
of stable complexes [log KLaL = 17.82(1)
and log KYbL = 19.65(1)]. Luminescence
studies on the Eu3+ and Tb3+ analogues evidenced
rather high emission quantum yields of 3.4 and 11%, respectively.
The emission lifetimes recorded in H2O and D2O solutions indicate the presence of a water molecule coordinated
to the metal ion. 1H nuclear magnetic relaxation dispersion
profiles and 17O NMR chemical shift and relaxation measurements
point to a rather low water exchange rate of the coordinated water
molecule (kex298 = 1.58 ×
106 s–1) and relatively high relaxivities
of 5.6 and 4.5 mM–1 s–1 at 20
MHz and 25 and 37 °C, respectively. Density functional theory
calculations and analysis of the paramagnetic shifts induced by Yb3+ indicate that the complexes adopt an unprecedented cis geometry
with the two picolinate groups situated on the same side of the coordination
sphere. Dissociation kinetics experiments were conducted by investigating
the exchange reactions of LuL occurring with Cu2+. The
results confirmed the beneficial effect of the rigid cyclohexyl group
on the inertness of the Lu3+ complex. Complex dissociation
occurs following proton- and metal-assisted pathways. The latter is
relatively efficient at neutral pH, thanks to the formation of a heterodinuclear
hydroxo complex. A
non-macrocyclic ligand containing a rigid cyclohexyl spacer
forms thermodynamically stable complexes with the lanthanide(III)
ions in aqueous solution. The complexes also show remarkable kinetic
inertness, though a structural change facilitates dissociation through
the metal-assisted mechanism for the small lanthanides. The Gd(III)
complex displays a relatively high relaxivity due to the presence
of a water molecule coordinated to the metal ion, while the Eu(III)
and Tb(III) analogues display strong metal-centered luminescence.
Collapse
Affiliation(s)
- Fátima Lucio-Martínez
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Zoltán Garda
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1, H-4010 Debrecen, Hungary
| | - Balázs Váradi
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1, H-4010 Debrecen, Hungary.,Doctoral School of Chemistry, University of Debrecen, Egyetem tér 1, H-4010 Debrecen, Hungary
| | - Ferenc Krisztián Kálmán
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1, H-4010 Debrecen, Hungary
| | - David Esteban-Gómez
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Éva Tóth
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d'Orléans, rue Charles Sadron, 45071 Orléans, Cedex 2, France
| | - Gyula Tircsó
- Department of Physical Chemistry, University of Debrecen, Egyetem tér 1, H-4010 Debrecen, Hungary
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| |
Collapse
|
7
|
Ingham A, Kostelnik TI, McNeil BL, Patrick BO, Choudhary N, Jaraquemada-Peláez MDG, Orvig C. Getting a lead on Pb 2+-amide chelators for 203/212Pb radiopharmaceuticals. Dalton Trans 2021; 50:11579-11595. [PMID: 34352061 DOI: 10.1039/d1dt01653a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Amide-based chelators DTPAm, EGTAm and ampam were synthesized to investigate which chelator most ideally coordinates [nat/203Pb]Pb2+ ions for potential radiopharmaceutical applications. 1H NMR spectroscopy was used to study each metal-ligand complex in the solution state. The 1H NMR spectrum of [Pb(DTPAm)]2+ revealed minimal isomerization and fluxional behaviour compared to [Pb(EGTAm)]2+ and [Pb(ampam)]2+, both of which showed fewer spectral changes indicative of less static behaviour. The solid-state coordination properties of each complex were also examined from single crystal structures that were studied by X-ray diffraction (XRD). In the solid-state, octadentate DTPAm coordinated Pb2+ to form an eight-coordinate hemidirected complex; octadentate EGTAm coordinated Pb2+ forming a ten-coordinate holodirected complex with a bidentate NO3- ion also coordinated to the metal centre; decadentate ampam completely encapsulated the Pb2+ ion to form a ten-coordinate holodirected complex with a C2 axis of symmetry. Potentiometric titrations were carried out to assess the thermodynamic stability of each metal-ligand complex. The pM values obtained for [Pb(DTPAm)]2+, [Pb(EGTAm)]2+ and [Pb(ampam)]2+ were 9.7, 7.2 and 10.2, respectively. The affinity of each chelator for Pb2+ ions was tested by [203Pb]Pb2+ radiolabeling studies to evaluate their prospects as chelators for [203/212Pb]Pb2+-based radiopharmaceuticals. DTPAm radiolabeled [203Pb]Pb2+ ions achieving molar activities as high as 3.5 MBq μmol-1 within 15 minutes, at 25 °C, whereas EGTAm and ampam produced lower molar activities of 0.25 MBq μmol-1 within 30 minutes, at 37 °C. EGTAm and ampam were therefore deemed unsuitable for [203/212Pb]Pb2+-based radiopharmaceutical applications, while DTPAm warrants further studies.
Collapse
Affiliation(s)
- Aidan Ingham
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6 T 1Z1, Canada.
| | | | | | | | | | | | | |
Collapse
|
8
|
Sharma SK, Glaser JM, Edwards KJ, Sarbisheh EK, Salih AK, Lewis JS, Price EW. A Systematic Evaluation of Antibody Modification and 89Zr-Radiolabeling for Optimized Immuno-PET. Bioconjug Chem 2021; 32:1177-1191. [PMID: 32197571 PMCID: PMC9423892 DOI: 10.1021/acs.bioconjchem.0c00087] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Immuno-PET using desferrioxamine (DFO)-conjugated zirconium-89 ([89Zr]Zr4+)-labeled antibodies is a powerful tool used for preclinical and clinical molecular imaging. However, a comprehensive study evaluating the variables involved in DFO-conjugation and 89Zr-radiolabeling of antibodies and their impact on the in vitro and in vivo behavior of the resulting radioimmunoconjugates has not been adequately performed. Here, we synthesized different DFO-conjugates of the HER2-targeting antibody (Ab)-trastuzumab, dubbed T5, T10, T20, T60, and T200-to indicate the molar equivalents of DFO used for bioconjugation. Next we radiolabeled the immunoconjugates with ([89Zr]Zr4+) under a comprehensive set of reaction conditions including different buffers (PBS, chelexed-PBS, TRIS/HCl, HEPES; ± radioprotectants), different reaction volumes (0.1-1 mL), variable amounts of DFO-conjugated Ab (5, 25, 50 μg), and radioactivity (0.2-1.0 mCi; 7.4-37 MBq). We evaluated the effects of these variables on radiochemical yield (RCY), molar activity (Am)/specific activity (As), immunoreactive fraction, and ultimately the in vivo biodistribution profile and tumor targeting ability of the trastuzumab radioimmunoconjugates. We show that increasing the degree of DFO conjugation to trastuzumab increased the RCY (∼90%) and Am/As (∼194 MBq/nmol; 35 mCi/mg) but decreased the HER2-binding affinity (3.5×-4.6×) and the immunoreactive fraction of trastuzumab down to 50-64%, which translated to dramatically inferior in vivo performance of the radioimmunoconjugate. Cell-based immunoreactivity assays and standard binding affinity analyses using surface plasmon resonance (SPR) did not predict the poor in vivo performance of the most extreme T200 conjugate. However, SPR-based concentration free calibration analysis yielded active antibody concentration and was predictive of the in vivo trends. Positron emission tomography (PET) imaging and biodistribution studies in a HER2-positive xenograft model revealed activity concentrations of 38.7 ± 3.8 %ID/g in the tumor and 6.3 ± 4.1 %ID/g in the liver for ([89Zr]Zr4+)-T5 (∼1.4 ± 0.5 DFOs/Ab) at 120 h after injection of the radioimmunoconjugates. On the other hand, ([89Zr]Zr4+)-T200 (10.9 ± 0.7 DFOs/Ab) yielded 16.2 ± 3.2 %ID/g in the tumor versus 27.5 ± 4.1 %ID/g in the liver. Collectively, our findings suggest that synthesizing trastuzumab immunoconjugates bearing 1-3 DFOs per Ab (T5 and T10) combined with radiolabeling performed in low reaction volumes using Chelex treated PBS or HEPEs without a radioprotectant provided radioimmunoconjugates having high Am/As (97 MBq/nmol; 17.5 ± 2.2 mCi/mg), highly preserved immunoreactive fractions (86-93%), and favorable in vivo biodistribution profile with excellent tumor uptake.
Collapse
Affiliation(s)
- Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jonathan M. Glaser
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Kimberly J. Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Akam K. Salih
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK, S7N-5C9, Canada
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Eric W. Price
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK, S7N-5C9, Canada
| |
Collapse
|
9
|
Klasen B, Moon ES, Rösch F. AAZTA 5-squaramide ester competing with DOTA-, DTPA- and CHX-A″-DTPA-analogues: Promising tool for 177Lu-labeling of monoclonal antibodies under mild conditions. Nucl Med Biol 2021; 96-97:80-93. [PMID: 33839678 DOI: 10.1016/j.nucmedbio.2021.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/12/2021] [Accepted: 03/20/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Combining the advantages of both cyclic and acyclic chelator systems, AAZTA (1,4-bis(carboxymethyl)-6-[bis(carboxymethyl)]amino-6-methylperhydro-1,4-diazepine) is well suited for complexation of various diagnostic and therapeutic radiometals such as gallium-68, scandium-44 and lutetium-177 under mild conditions. Due to its specificity for primary amines and pH dependent binding properties, squaric acid (SA) represents an excellent tool for selective coupling of the appropriate chelator to different target vectors. Therefore, the aim of this study was to evaluate radiolabeling properties of the novel bifunctional AAZTA5-SA being coupled to a model antibody (bevacizumab) in comparison to DOTA-SA, DTPA-p-Bn-SA and CHX-A″-DTPA-p-Bn-SA using the therapeutic nuclide lutetium-177. METHODS AND RESULTS As proof-of-concept, bevacizumab was first functionalized with AAZTA5-SA, DOTA-SA, DTPA-p-Bn-SA or CHX-A″-DTPA-p-Bn-SA. After purification via fractionated size exclusion chromatography (SEC), the corresponding immunoconjugates were subsequently radiolabeled with lutetium-177 at pH 7 and room temperature (RT) as well as 37 °C. After 90 min, labeling of AAZTA5-SA-mAb resulted in almost quantitative radiochemical yields (RCY) of >98% and >99%, respectively. Formation of [177Lu]Lu-DTPA-p-Bn-SA-mAb indicated rapid labeling kinetics reaching similar yields at RT already after 30 min. Fast but incomplete radiolabeling of the CHX-A″-analogue could be observed with a yield of 74% after 10 min and no further significant increase. In contrast, 177Lu-labeling of DOTA-SA-mAb showed negligible radiochemical yields of <2% both at room temperature and 37 °C. In vitro complex stability measurements of [177Lu]Lu-AAZTA5-SA-mAb at 37 °C indicated >94% protein bound activity in human serum and >92% in phosphate buffered saline (PBS), respectively within 15 days. [177Lu]Lu-DTPA-p-Bn-SA-mAb and [177Lu]Lu-CHX-A″-DTPA-p-Bn-SA-mAb revealed similar to even slightly higher in vitro stability in both media. CONCLUSION Coupling of AAZTA5-SA to the monoclonal antibody bevacizumab allowed for 177Lu-labeling with almost quantitative radiochemical yields both at room temperature and 37 °C. Within 15 days, the resulting radioconjugate indicated very high in vitro complex stability both in human serum and PBS. Therefore, AAZTA5-SA is a promising tool for 177Lu-labeling of sensitive biomolecules such as antibodies for theranostic applications.
Collapse
Affiliation(s)
- Benedikt Klasen
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany.
| | - Euy Sung Moon
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany.
| | - Frank Rösch
- Department of Chemistry - TRIGA site, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
10
|
Herrero Álvarez N, Bauer D, Hernández-Gil J, Lewis JS. Recent Advances in Radiometals for Combined Imaging and Therapy in Cancer. ChemMedChem 2021; 16:2909-2941. [PMID: 33792195 DOI: 10.1002/cmdc.202100135] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Indexed: 12/14/2022]
Abstract
Nuclear medicine is defined as the use of radionuclides for diagnostic and therapeutic applications. The imaging modalities positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are based on γ-emissions of specific energies. The therapeutic technologies are based on β- -particle-, α-particle-, and Auger electron emitters. In oncology, PET and SPECT are used to detect cancer lesions, to determine dosimetry, and to monitor therapy effectiveness. In contrast, radiotherapy is designed to irreparably damage tumor cells in order to eradicate or control the disease's progression. Radiometals are being explored for the development of diagnostic and therapeutic radiopharmaceuticals. Strategies that combine both modalities (diagnostic and therapeutic), referred to as theranostics, are promising candidates for clinical applications. This review provides an overview of the basic concepts behind therapeutic and diagnostic radiopharmaceuticals and their significance in contemporary oncology. Select radiometals that significantly impact current and upcoming cancer treatment strategies are grouped as clinically suitable theranostics pairs. The most important physical and chemical properties are discussed. Standard production methods and current radionuclide availability are provided to indicate whether a cost-efficient use in a clinical routine is feasible. Recent preclinical and clinical developments and outline perspectives for the radiometals are highlighted in each section.
Collapse
Affiliation(s)
- Natalia Herrero Álvarez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - David Bauer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Javier Hernández-Gil
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Katholieke Universiteit, Herestraat 49, 3000, Leuven, Belgium
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.,Department of Pharmacology, Weill-Cornell Medical College, New York, NY, 10065, USA
| |
Collapse
|
11
|
Dewulf J, Adhikari K, Vangestel C, Wyngaert TVD, Elvas F. Development of Antibody Immuno-PET/SPECT Radiopharmaceuticals for Imaging of Oncological Disorders-An Update. Cancers (Basel) 2020; 12:E1868. [PMID: 32664521 PMCID: PMC7408676 DOI: 10.3390/cancers12071868] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 01/12/2023] Open
Abstract
Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are molecular imaging strategies that typically use radioactively labeled ligands to selectively visualize molecular targets. The nanomolar sensitivity of PET and SPECT combined with the high specificity and affinity of monoclonal antibodies have shown great potential in oncology imaging. Over the past decades a wide range of radio-isotopes have been developed into immuno-SPECT/PET imaging agents, made possible by novel conjugation strategies (e.g., site-specific labeling, click chemistry) and optimization and development of novel radiochemistry procedures. In addition, new strategies such as pretargeting and the use of antibody fragments have entered the field of immuno-PET/SPECT expanding the range of imaging applications. Non-invasive imaging techniques revealing tumor antigen biodistribution, expression and heterogeneity have the potential to contribute to disease diagnosis, therapy selection, patient stratification and therapy response prediction achieving personalized treatments for each patient and therefore assisting in clinical decision making.
Collapse
Affiliation(s)
- Jonatan Dewulf
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Karuna Adhikari
- Faculty of Pharmaceutical Biomedical and Veterinary Sciences, Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium;
| | - Christel Vangestel
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Tim Van Den Wyngaert
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Filipe Elvas
- Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium; (J.D.); (C.V.); (T.V.D.W.)
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, B-2650 Edegem, Belgium
| |
Collapse
|
12
|
Li L, Kuo HT, Wang X, Merkens H, Colpo N, Radchenko V, Schaffer P, Lin KS, Bénard F, Orvig C. tBu 4octapa-alkyl-NHS for metalloradiopeptide preparation. Dalton Trans 2020; 49:7605-7619. [PMID: 32459231 DOI: 10.1039/d0dt00845a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The peptide is an important class of biological targeting molecule; herein, a new bifunctional octadentate non-macrocyclic H4octapa, tBu4octapa-alkyl-NHS, which is compatible with solid-phase peptide synthesis and thus useful for radiopeptide preparation, has been synthesized. To preserve denticity, the alkyl-N-hydroxylsuccinimide linker was covalently attached to the methylene-carbon on one of the acetate arms, yielding a chiral carbon center. According to density-functional theory (DFT) calculations using [Lu(octapa-alkyl-benzyl-ester)]- as a simulation model, the chirality has minimal effects on the complex geometry; regardless of the S-/R-stereochemistry, DFT calculations revealed two possible geometric isomers, distorted bicapped trigonal antiprism (DBTA) and distorted square antiprism (DSA), due to the asymmetry in the chelator. To evaluate the biological behavior of the new bifunctionalization, two well-studied PSMA (prostate-specific membrane antigen)-targeting peptidomimetics of varying hydrophobicity were chosen as proof-of-principle targeting vector molecules. Radiolabeling both bioconjugates with lutetium-177 was highly efficient at room temperature in 15 min at micromolar chelator concentration pH = 7. Both the in vitro serum challenge and the lanthanum(iii) challenge studies revealed complex lability, and notably, progressive bone accumulation was only observed with the more hydrophobic linker (i.e. H4octapa-alkyl-PSMA617). This in vivo result informs potential alterations exerted by the linker on the complex geometry and stability, with an appropriate biological targeting vector adopted for such evaluations.
Collapse
Affiliation(s)
- Lily Li
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada. and Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Hsiou-Ting Kuo
- Department of Molecular Oncology, BC Cancer, 675 West 10th Ave, Vancouver, British Columbia V5Z 1L3, Canada
| | - Xiaozhu Wang
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer, 675 West 10th Ave, Vancouver, British Columbia V5Z 1L3, Canada
| | - Nadine Colpo
- Department of Molecular Oncology, BC Cancer, 675 West 10th Ave, Vancouver, British Columbia V5Z 1L3, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada and Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6 T 1Z1, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, 675 West 10th Ave, Vancouver, British Columbia V5Z 1L3, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, 675 West 10th Ave, Vancouver, British Columbia V5Z 1L3, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| |
Collapse
|
13
|
Li L, de Guadalupe Jaraquemada-Peláez M, Aluicio-Sarduy E, Wang X, Barnhart TE, Cai W, Radchenko V, Schaffer P, Engle JW, Orvig C. Coordination chemistry of [Y(pypa)] - and comparison immuno-PET imaging of [ 44Sc]Sc- and [ 86Y]Y-pypa-phenyl-TRC105. Dalton Trans 2020; 49:5547-5562. [PMID: 32270167 PMCID: PMC7222037 DOI: 10.1039/d0dt00437e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both scandium-44 and yttrium-86 are popular PET isotopes with appropriate half-lives for immuno-positron emission tomography (immuno-PET) imaging. Herein, a new bifunctional H4pypa ligand, H4pypa-phenyl-NCS, is synthesized, conjugated to a monoclonal antibody, TRC105, and labeled with both radionuclides to investigate the long-term in vivo stability of each complex. While the 44Sc-labeled radiotracer exhibited promising pharmacokinetics and stability in 4T1-xenograft mice (n = 3) even upon prolonged interactions with blood serum proteins, the progressive bone uptake of the 86Y-counterpart indicated in vivo demetallation, obviating H4pypa as a suitable chelator for Y3+ ion in vivo. The solution chemistry of [natY(pypa)]- was studied in detail and the complex found to be thermodynamically stable in solution with a pM value 22.0, ≥3 units higher than those of the analogous DOTA- and CHX-A''-DTPA-complexes; the 86Y-result in vivo was therefore most unexpected. To explore further this in vivo lability, Density Functional Theory (DFT) calculation was performed to predict the geometry of [Y(pypa)]- and the results were compared with those for the analogous Sc- and Lu-complexes; all three adopted the same coordination geometry (i.e. distorted capped square antiprism), but the metal-ligand bonds were much longer in [Y(pypa)]- than in [Lu(pypa)]- and [Sc(pypa)]-, which could indicate that the size of the binding cavity is too small for the Y3+ ion, but suitable for both the Lu3+ and Sc3+ ions. Considered along with results from [86Y][Y(pypa-phenyl-TRC105)], it is noted that when matching chelators with radionuclides, chemical data such as the thermodynamic stability and in vitro inertness, albeit useful and necessary, do not always translate to in vivo inertness, especially with the prolonged blood circulation of the radiotracer bound to a monoclonal antibody. Although H4pypa is a nonadentate chelator, which theoretically matches the coordination number of the Y3+ ion, we show herein that its binding cavity, in fact, favors smaller metal ions such as Sc3+ and Lu3+ and further exploitation of the Sc-pypa combination is desired.
Collapse
Affiliation(s)
- Lily Li
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Hu A, Keresztes I, MacMillan SN, Yang Y, Ding E, Zipfel WR, DiStasio RA, Babich JW, Wilson JJ. Oxyaapa: A Picolinate-Based Ligand with Five Oxygen Donors that Strongly Chelates Lanthanides. Inorg Chem 2020; 59:5116-5132. [PMID: 32216281 DOI: 10.1021/acs.inorgchem.0c00372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Coordination compounds of the lanthanide ions (Ln3+) have important applications in medicine due to their photophysical, magnetic, and nuclear properties. To effectively use the Ln3+ ions for these applications, chelators that stably bind them in vivo are required to prevent toxic side effects that arise from localization of these ions in off-target tissue. In this study, two new picolinate-containing chelators, a heptadentate ligand OxyMepa and a nonadentate ligand Oxyaapa, were prepared, and their coordination chemistries with Ln3+ ions were thoroughly investigated to evaluate their suitability for use in medicine. Protonation constants of these chelators and stability constants for their Ln3+ complexes were evaluated. Both ligands exhibit a thermodynamic preference for small Ln3+ ions. The log KLuL = 12.21 and 21.49 for OxyMepa and Oxyaapa, respectively, indicating that the nonadentate Oxyaapa forms complexes of significantly higher stability than the heptadentate OxyMepa. X-ray crystal structures of the Lu3+ complexes were obtained, revealing that Oxyaapa saturates the coordination sphere of Lu3+, whereas OxyMepa leaves an additional open coordination site for a bound water ligand. Solution structural studies carried out with NMR spectroscopy revealed the presence of two possible conformations for these ligands upon Ln3+ binding. Density functional theory (DFT) calculations were applied to probe the geometries and energies of these conformations. Energy differences obtained by DFT are small but consistent with experimental data. The photophysical properties of the Eu3+ and Tb3+ complexes were characterized, revealing modest photoluminescent quantum yields of <2%. Luminescence lifetime measurements were carried out in H2O and D2O, showing that the Eu3+ and Tb3+ complexes of OxyMepa have two inner-sphere water ligands, whereas the Eu3+ and Tb3+ complexes of Oxyaapa have zero. Lastly, variable-temperature 17O NMR spectroscopy was performed for the Gd-OxyMepa complex to determine its water exchange rate constant of kex298 = (2.8 ± 0.1) × 106 s-1. Collectively, this comprehensive characterization of these Ln3+ chelators provides valuable insight for their potential use in medicine and garners additional understanding of ligand design strategies.
Collapse
Affiliation(s)
- Aohan Hu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Ivan Keresztes
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Yang Yang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Erdong Ding
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Warren R Zipfel
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Robert A DiStasio
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - John W Babich
- Department of Radiology, Weill Cornell Medicine, New York, New York 10065, United States
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
15
|
Tickner BJ, Stasiuk GJ, Duckett SB, Angelovski G. The use of yttrium in medical imaging and therapy: historical background and future perspectives. Chem Soc Rev 2020; 49:6169-6185. [DOI: 10.1039/c9cs00840c] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Yttrium presents a wide palette of isotopes with interesting coordination and radiochemical properties. We review its most prominent isotopes and their diverse medical uses in therapy and imaging.
Collapse
Affiliation(s)
- Ben J. Tickner
- Centre for Hyperpolarisation in Magnetic Resonance
- Department of Chemistry
- University of York
- Heslington
- UK
| | - Graeme J. Stasiuk
- Department of Imaging Chemistry and Biology
- School of Biomedical Engineering and Imaging
- King's College London
- London
- UK
| | - Simon B. Duckett
- Centre for Hyperpolarisation in Magnetic Resonance
- Department of Chemistry
- University of York
- Heslington
- UK
| | - Goran Angelovski
- MR Neuroimaging Agents
- Max Planck Institute for Biological Cybernetics
- Tuebingen
- Germany
| |
Collapse
|
16
|
Pandey U, Kameswaran M, Gamre N, Dash A. Preparation of 177 Lu-labeled Nimotuzumab for radioimmunotherapy of EGFR-positive cancers: Comparison of DOTA and CHX-A″-DTPA as bifunctional chelators. J Labelled Comp Radiopharm 2019; 62:158-165. [PMID: 30663095 DOI: 10.1002/jlcr.3707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/11/2019] [Indexed: 11/05/2022]
Abstract
This study was aimed at evaluating the role of bifunctional chelators DOTA-NCS and CHX-A″-DTPA-NCS used for conjugating 177 Lu with Nimotuzumab on the radiochemical yields, purity, in vitro stability, and specificity of the radioimmunoconjugates to EGFR. Two immunoconjugates were prepared wherein Nimotuzumab was conjugated with the acyclic ligand p-NCS-Bn-CHX-A″-DTPA and macrocyclic ligand p-NCS-Bn-DOTA. These were radiolabeled with 177 Lu, purified on PD-10 column, and characterized by SE-HPLC. In vitro stability was determined up to 4 days post preparation. Specificity of the radioimmunoconjugates was ascertained by in vitro studies in A431 cells while the biodistribution patterns were studied in normal Swiss mice up to 96 hours post injection. Four to five molecules of CHX-A″-DTPA/DOTA were attached to one molecule of Nimotuzumab. Radiochemical purity of both 177 Lu-CHX-A″-DTPA-Nimotuzumab and 177 Lu-DOTA-Nimotuzumab was determined to be greater than 98%. Both the radioimmunoconjugates exhibited good in vitro stability at 37°C up to 4 days post preparation in saline, and their clearance was largely by the hepatobiliary route. The DOTA- and CHX-A″-DTPA-based radioimmunoconjugates could be prepared with good radiochemical purity, in vitro stability, and specificity to EGFR. Further studies in EGFR-positive cancers would pave way for them for use in the clinics.
Collapse
Affiliation(s)
- Usha Pandey
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Bhabha Atomic Research Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Mythili Kameswaran
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Naresh Gamre
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India.,Bhabha Atomic Research Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
17
|
Basaco T, Pektor S, Bermudez JM, Meneses N, Heller M, Galván JA, Boligán KF, Schürch S, von Gunten S, Türler A, Miederer M. Evaluation of Radiolabeled Girentuximab In Vitro and In Vivo. Pharmaceuticals (Basel) 2018; 11:E132. [PMID: 30487460 PMCID: PMC6316122 DOI: 10.3390/ph11040132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 11/16/2022] Open
Abstract
Girentuximab (cG250) targets carbonic anhydrase IX (CAIX), a protein which is expressed on the surface of most renal cancer cells (RCCs). cG250 labeled with 177Lu has been used in clinical trials for radioimmunotherapy (RIT) of RCCs. In this work, an extensive characterization of the immunoconjugates allowed optimization of the labeling conditions with 177Lu while maintaining immunoreactivity of cG250, which was then investigated in in vitro and in vivo experiments. cG250 was conjugated with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (DOTA(SCN)) by using incubation times between 30 and 90 min and characterized by mass spectrometry. Immunoconjugates with five to ten DOTA(SCN) molecules per cG250 molecule were obtained. Conjugates with ratios less than six DOTA(SCN)/cG250 had higher in vitro antigen affinity, both pre- and postlabeling with 177Lu. Radiochemical stability increased, in the presence of sodium ascorbate, which prevents radiolysis. The immunoreactivity of the radiolabeled cG250 tested by specific binding to SK-RC-52 cells decreased when the DOTA content per conjugate increased. The in vivo tumor uptake was < 10% ID/g and independent of the total amount of protein in the range between 5 and 100 µg cG250 per animal. Low tumor uptake was found to be due to significant necrotic areas and heterogeneous CAIX expression. In addition, low vascularity indicated relatively poor accessibility of the CAIX target.
Collapse
Affiliation(s)
- Tais Basaco
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
- Laboratory of Radiochemistry, Paul Scherrer Institute (PSI), 5232 Villigen PSI, Switzerland.
| | - Stefanie Pektor
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| | - Josue M Bermudez
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Niurka Meneses
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Manfred Heller
- Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland.
| | - José A Galván
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland.
| | - Kayluz F Boligán
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Stephan von Gunten
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Andreas Türler
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Matthias Miederer
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| |
Collapse
|
18
|
Abstract
Radiometals possess an exceptional breadth of decay properties and have been applied to medicine with great success for several decades. The majority of current clinical use involves diagnostic procedures, which use either positron-emission tomography (PET) or single-photon imaging to detect anatomic abnormalities that are difficult to visualize using conventional imaging techniques (e.g., MRI and X-ray). The potential of therapeutic radiometals has more recently been realized and relies on ionizing radiation to induce irreversible DNA damage, resulting in cell death. In both cases, radiopharmaceutical development has been largely geared toward the field of oncology; thus, selective tumor targeting is often essential for efficacious drug use. To this end, the rational design of four-component radiopharmaceuticals has become popularized. This Review introduces fundamental concepts of drug design and applications, with particular emphasis on bifunctional chelators (BFCs), which ensure secure consolidation of the radiometal and targeting vector and are integral for optimal drug performance. Also presented are detailed accounts of production, chelation chemistry, and biological use of selected main group and rare earth radiometals.
Collapse
Affiliation(s)
- Thomas I Kostelnik
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| |
Collapse
|
19
|
Boros E, Holland JP. Chemical aspects of metal ion chelation in the synthesis and application antibody-based radiotracers. J Labelled Comp Radiopharm 2018; 61:652-671. [PMID: 29230857 PMCID: PMC5997514 DOI: 10.1002/jlcr.3590] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
Abstract
Radiometals are becoming increasingly accessible and are utilized frequently in the design of radiotracers for imaging and therapy. Nuclear properties ranging from the emission of γ-rays and β+ -particles (imaging) to Auger electron and β- and α-particles (therapy) in combination with long half-lives are ideally matched with the relatively long biological half-life of monoclonal antibodies in vivo. Radiometal labeling of antibodies requires the incorporation of a metal chelate onto the monoclonal antibody. This chelate must coordinate the metal under mild conditions required for the handling of antibodies, as well as provide high kinetic, thermodynamic, and metabolic stability once the metal ion is coordinated to prevent release of the radionuclide before the target site is reached in vivo. Herein, we review the role of different radiometals that have found applications of the design of radiolabeled antibodies for imaging and radioimmunotherapy. Each radionuclide is described regarding its nuclear synthesis, coordinative preference, and radiolabeling properties with commonly used and novel chelates, as well as examples of their preclinical and clinical applications. An overview of recent trends in antibody-based radiopharmaceuticals is provided to spur continued development of the chemistry and application of radiometals for imaging and therapy.
Collapse
Affiliation(s)
- Eszter Boros
- Stony Brook University, Department of Chemistry, 100 Nicolls road, 11790 Stony Brook, NY, United States
| | - Jason P. Holland
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
20
|
Massicano AVF, Marquez-Nostra BV, Lapi SE. Targeting HER2 in Nuclear Medicine for Imaging and Therapy. Mol Imaging 2018; 17:1536012117745386. [PMID: 29357745 PMCID: PMC5784567 DOI: 10.1177/1536012117745386] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/17/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022] Open
Abstract
Since its discovery, the human epidermal growth factor 2 (HER2) has been extensively studied. Presently, there are 2 standard diagnostic techniques to assess HER2 status in biopsies: immunohistochemistry and fluorescence in situ hybridization. While these techniques have played an important role in the treatment of patients with HER2-positive cancer, they both require invasive biopsies for analysis. Moreover, the expression of HER2 is heterogeneous in breast cancer and can change over the course of the disease. Thus, the degree of HER2 expression in the small sample size of biopsied tumors at the time of analysis may not represent the overall status of HER2 expression in the whole tumor and in between tumor foci in the metastatic setting as the disease progresses. Unlike biopsy, molecular imaging using probes against HER2 allows for a noninvasive, whole-body assessment of HER2 status in real time. This technique could potentially select patients who may benefit from HER2-directed therapy and offer alternative treatments to those who may not benefit. Several antibodies and small molecules against HER2 have been labeled with different radioisotopes for nuclear imaging and/or therapy. This review presents the most recent advances in HER2 targeting in nuclear medicine focusing on preclinical and clinical studies.
Collapse
Affiliation(s)
| | | | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|