1
|
Tran HH, Yamaguchi A, Manning HC. Radiotheranostic landscape: A review of clinical and preclinical development. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07103-7. [PMID: 39891713 DOI: 10.1007/s00259-025-07103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Radiotheranostics combines diagnostic imaging with targeted radionuclide therapy, representing a transformative approach in precision oncology. Landmark approvals of Lutathera® and Pluvicto® have catalyzed significant advancements in this field, driving research into novel radionuclides, targeting strategies, and clinical applications. This review evaluates the evolving clinical and preclinical landscape of radiotheranostics, highlighting advancements, emerging trends, and persistent challenges in radionuclide therapy. METHODS A comprehensive analysis was performed, encompassing active clinical trials as of December 2024, sourced from ClinicalTrials.gov and TheranosticTrials.org. Preclinical developments were evaluated through a review of recent literature, focusing on innovations in radionuclide production, targeting molecules, and radiochemistry. RESULTS In reviewing the clinical landscape, agents targeting somatostatin receptors (SSTR) and prostate-specific membrane antigen (PSMA) still dominate the field, but new targets such as fibroblast activation protein (FAP), integrins, and gastrin-releasing peptide receptors (GRPR) are gaining traction in both clinical and preclinical development. While small molecules and peptides remain the most common radionuclide carriers, antibody-based carriers including bispecific antibodies, immunoglobin-derived antigen-binding fragments, and antibody-mimetic proteins are on the rise due to their specificity and adaptability. Innovations in radioligand design are driving a shift from agonists to antagonists, accompanied by the development of modified peptides with enhanced pharmacokinetics and tumor-targeting properties. Next-generation therapeutic radionuclides, such as the beta-emitter terbium-161 and alpha-emitters actinium-225 and lead-212, are under investigation to complement or replace lutetium-177, addressing the need for improved efficacy and reduced toxicity. Paired isotopic radionuclides are gaining popularity for their ability to optimize imaging and therapeutic dosimetry as they offer near-identical specificity, biodistribution, and metabolism. Additionally, radiohybrid systems represent an innovative approach to chelating chemically distinct radionuclide pairs within a single molecule, further enhancing flexibility in radiotheranostic design. CONCLUSION Radiotheranostics has transformed cancer care through its precision and adaptability, but challenges in radionuclide production, regulatory frameworks, and workforce training hinder broader adoption. Advances in isotopic pairing, next-generation radionuclides, and radiohybrid systems in preclinical and clinical settings hold promise to overcome these barriers. Collaborative efforts among academia, industry, and regulatory bodies are critical to accelerating innovation and optimizing clinical outcomes.
Collapse
Affiliation(s)
- Ha H Tran
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aiko Yamaguchi
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Kurakina ES, McNeil BL, Khushvaktov J, Temerbulatova NT, Mirzayev NA, Magomedbekov EP, Hoehr C, Ramogida CF, Filosofov DV, Radchenko V. Production and purification of radiolabeling-ready 132/135La from the irradiation of metallic natBa targets with low energy protons. Nucl Med Biol 2025; 144-145:108994. [PMID: 39889329 DOI: 10.1016/j.nucmedbio.2025.108994] [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: 10/17/2024] [Revised: 12/26/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
INTRODUCTION Radiolanthanides 132La and 135La form a promising chemically matched theranostic pair. With a half-life of 18.95 h, 135La acts as the therapeutic isotope as it releases approximately 11 Auger electrons per decay, making it compatible with targeted Auger electron therapy (TAET), whereas 132La with half-life of 4.58 h undergoes positron emission making it compatible with imaging via positron emission tomography (PET). METHODS 132/135La were produced via irradiation of natural barium targets (99.9 %) with 12.8 MeV protons. A two-step separation scheme using extraction chromatographic resin TK200 (50-100 μm) and cation exchange resin Dowex 50Wx4 (200-400 mesh) was designed. Inductively coupled plasma mass spectrometry (ICP-MS) was used to quantify non-radioactive impurities in each fraction of the separation method. The distribution coefficients of La3+ in HNO3 on the TK200 resin and on both Dowex 50Wx8 (200-400 mesh) and Dowex 50Wx4 resins in ammonium α-hydroxyisobutyrate (pH 4.8) were determined, respectively. RESULTS This novel separation scheme allowed for reliable separation of [132/135La]La3+ from the Ba2+ target material, resulting in a high radiochemical yield of 98.3 ± 2.1 % (n = 3) with the final elute being directly compatible with subsequent radiolabeling due to the use of ammonium α-hydroxyisobutyrate to eliminate steps in the radiopharmaceutical synthetic process.
Collapse
Affiliation(s)
- E S Kurakina
- Dzhelepov Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, Dubna 141980, Russian Federation; Department of High-Energy Chemistry and Radioecology, D. Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russian Federation
| | - B L McNeil
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
| | - J Khushvaktov
- Dzhelepov Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, Dubna 141980, Russian Federation
| | - N T Temerbulatova
- Dzhelepov Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, Dubna 141980, Russian Federation
| | - N A Mirzayev
- Dzhelepov Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, Dubna 141980, Russian Federation; Institute of Radiation Problems, Ministry of Science and Education Republic of Azerbaijan, Baku АZ1143, Azerbaijan; Khazar University, 13 Mahsati 41, Baku AZ1096, Azerbaijan
| | - E P Magomedbekov
- Department of High-Energy Chemistry and Radioecology, D. Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russian Federation
| | - C Hoehr
- Department of Physics and Astronomy, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada; Department of Computer Science, Mathematics, Physics, and Statistics, University of British Columbia Okanagan, Kelowna, British Columbia V1V 1V7, Canada
| | - C F Ramogida
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
| | - D V Filosofov
- Dzhelepov Laboratory of Nuclear Problems, Joint Institute for Nuclear Research, Dubna 141980, Russian Federation
| | - V Radchenko
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.
| |
Collapse
|
3
|
Pandey A, Rohweder PJ, Chan LM, Ongpipattanakul C, Chung DH, Paolella B, Quimby FM, Nguyen N, Verba KA, Evans MJ, Craik CS. Therapeutic Targeting and Structural Characterization of a Sotorasib-Modified KRAS G12C-MHC I Complex Demonstrate the Antitumor Efficacy of Hapten-Based Strategies. Cancer Res 2025; 85:329-341. [PMID: 39656104 PMCID: PMC11733532 DOI: 10.1158/0008-5472.can-24-2450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/11/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025]
Abstract
Antibody-based therapies have emerged as a powerful strategy for the management of diverse cancers. Unfortunately, tumor-specific antigens remain challenging to identify and target. Recent work established that inhibitor-modified peptide adducts derived from KRAS G12C are competent for antigen presentation via MHC I and can be targeted by antibody-based therapeutics, offering a means to directly target an intracellular oncoprotein at the cell surface with combination therapies. Here, we validated the antigen display of "haptenated" KRAS G12C peptide fragments on tumors in mouse models treated with the FDA-approved KRAS G12C covalent inhibitor sotorasib using PET/CT imaging of an 89Zr-labeled P1B7 IgG antibody, which selectively binds sotorasib-modified KRAS G12C-MHC I complexes. Targeting this peptide-MHC I complex with radioligand therapy using 225Ac- or 177Lu-P1B7 IgG effectively inhibited tumor growth in combination with sotorasib. Elucidation of the 3.1 Å cryo-EM structure of P1B7 bound to a haptenated KRAS G12C peptide-MHC I complex confirmed that the sotorasib-modified KRAS G12C peptide is presented via a canonical binding pose and showed that P1B7 binds the complex in a T-cell receptor-like manner. Together, these findings demonstrate the potential value of targeting unique oncoprotein-derived, haptenated MHC I complexes with radioligand therapeutics and provide a structural framework for developing next generation antibodies. Significance: Radioligand therapy using an antibody targeting KRAS-derived, sotorasib-modified MHC I complexes elicits antitumor effects superior to those of sotorasib alone and provides a potential strategy to repurpose sotorasib as a hapten to overcome resistance.
Collapse
Affiliation(s)
- Apurva Pandey
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Peter J. Rohweder
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Lieza M. Chan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Chayanid Ongpipattanakul
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Dong hee Chung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Bryce Paolella
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Fiona M. Quimby
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Ngoc Nguyen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Kliment A. Verba
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California
| | - Michael J. Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| |
Collapse
|
4
|
Bauer D, De Gregorio R, Pratt EC, Bell A, Michel A, Lewis JS. Examination of the PET in vivo generator 134Ce as a theranostic match for 225Ac. Eur J Nucl Med Mol Imaging 2024; 51:4015-4025. [PMID: 38940841 DOI: 10.1007/s00259-024-06811-w] [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] [Received: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
PURPOSE The radionuclide pair cerium-134/lanthanum-134 (134Ce/134La) was recently proposed as a suitable diagnostic counterpart for the therapeutic alpha-emitter actinium-225 (225Ac). The unique properties of 134Ce offer perspectives for developing innovative in vivo investigations that are not possible with 225Ac. In this work, 225Ac- and 134Ce-labelled tracers were directly compared using internalizing and slow-internalizing cancer models to evaluate their in vivo comparability, progeny meandering, and potential as a matched theranostic pair for clinical translation. Despite being an excellent chemical match, 134Ce/134La has limitations to the setting of quantitative positron emission tomography imaging. METHODS The precursor PSMA-617 and a macropa-based tetrazine-conjugate (mcp-PEG8-Tz) were radiolabelled with 225Ac or 134Ce and compared in vitro and in vivo using standard (radio)chemical methods. Employing biodistribution studies and positron emission tomography (PET) imaging in athymic nude mice, the radiolabelled PSMA-617 tracers were evaluated in a PC3/PIP (PC3 engineered to express a high level of prostate-specific membrane antigen) prostate cancer mouse model. The 225Ac and 134Ce-labelled mcp-PEG8-Tz were investigated in a BxPC-3 pancreatic tumour model harnessing the pretargeting strategy based on a trans-cyclooctene-modified 5B1 monoclonal antibody. RESULTS In vitro and in vivo studies with both 225Ac and 134Ce-labelled tracers led to comparable results, confirming the matching pharmacokinetics of this theranostic pair. However, PET imaging of the 134Ce-labelled precursors indicated that quantification is highly dependent on tracer internalization due to the redistribution of 134Ce's PET-compatible daughter 134La. Consequently, radiotracers based on internalizing vectors like PSMA-617 are suited for this theranostic pair, while slow-internalizing 225Ac-labelled tracers are not quantitatively represented by 134Ce PET imaging. CONCLUSION When employing slow-internalizing vectors, 134Ce might not be an ideal match for 225Ac due to the underestimation of tumour uptake caused by the in vivo redistribution of 134La. However, this same characteristic makes it possible to estimate the redistribution of 225Ac's progeny noninvasively. In future studies, this unique PET in vivo generator will further be harnessed to study tracer internalization, trafficking of receptors, and the progression of the tumour microenvironment.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Roberto De Gregorio
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Edwin C Pratt
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Abram Bell
- Brigham Young University-Idaho, Rexburg, ID, 83440, USA
| | - Alexa Michel
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jason S Lewis
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Kanagasundaram T, Sun Y, Lee KK, MacMillan SN, Brugarolas P, Wilson JJ. Fluorine-18 incorporation and radiometal coordination in macropa ligands for PET imaging and targeted alpha therapy. Chem Commun (Camb) 2024; 60:11940-11943. [PMID: 39352495 DOI: 10.1039/d4cc04165h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
The development of theranostic agents for radiopharmaceuticals based on therapeutic alpha emitters marks an important clinical need. We describe a strategy for the development of theranostic agents of this type via the functionalization of the ligand with the diagnostic radionuclide fluorine-18. An analogue of macropa, an 18-membered macrocyclic chelator with high affinity for alpha therapeutic radiometals, was synthesized and its complexation properties with metal ions were determined. The new macropa-F ligand was used for quantitative radiometal complexation with lead-203 and bismuth-207, as surrogates for their alpha-emitting radioisotopes. As a diagnostic partner, a radiofluorinated macropa ligand was used for quantitative bismuth(III) and lead(II) complexation. All fluorine-18 and radiometal complexes are highly stable in human serum over several days. This study presents a new proof-of-principle approach for developing theranostic agents based on alpha-emitting radionuclides and fluorine-18.
Collapse
Affiliation(s)
- Thines Kanagasundaram
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
| | - Yang Sun
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA-02114, USA.
| | - Kevin K Lee
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA-93106, USA.
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
| | - Pedro Brugarolas
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA-02114, USA.
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA-93106, USA.
| |
Collapse
|
6
|
Lee KK, Chakraborty M, Hu A, Kanagasundaram T, Thorek DLJ, Wilson JJ. Chelation of [ 111In]In 3+ with the dual-size-selective macrocycles py-macrodipa and py 2-macrodipa. Dalton Trans 2024; 53:14634-14647. [PMID: 39163366 PMCID: PMC11663299 DOI: 10.1039/d4dt02146k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Indium-111 (111In) is a diagnostic radiometal that is important in nuclear medicine for single-photon emission computed tomography (SPECT). In order to apply this radiometal, it needs to be stably chelated and conjugated to a targeting vector that delivers it to diseased tissue. Identifying effective chelators that are capable of binding and retaining [111In]In3+in vivo is an important research area. In this study, two 18-membered macrocyclic chelators, py-macrodipa and py2-macrodipa, were investigated for their ability to form stable coordination complexes with In3+ and to be effectively radiolabeled with [111In]In3+. The In3+ complexes of these two chelators were characterized by NMR spectroscopy, X-ray crystallography, and density functional theory calculations. These studies show that both py-macrodipa and py2-macrodipa form 8-coordinate In3+ complexes and attain an asymmetric conformation, consistent with prior studies on this ligand class with small rare earth metal ions. Spectrophotometric titrations were carried out to determine the thermodynamic stability constants (log KML) of [In(py-macrodipa)]+ and [In(py2-macrodipa)]+, which were found to be 18.96(6) and 19.53(5), respectively, where the values in parentheses are the errors of the last significant figures obtained from the standard deviation from three independent replicates. Radiolabeling studies showed that py-macrodipa and py2-macrodipa can quantitatively be radiolabeled with [111In]In3+ at 25 °C within 5 min, even at ligand concentrations as low as 1 μM. The in vitro stability of the radiolabeled complexes was investigated in human serum at 37 °C, revealing that ∼90% of [111In][In(py-macrodipa)]+ and [111In][In(py2-macrodipa)]+ remained intact after 7 days. The biodistribution of these radiolabeled complexes in mice was investigated, showing lower uptake in the kidneys, liver, and blood at the 24 h mark compared to [111In]InCl3. These results demonstrate the potential of py-macrodipa and py2-macrodipa as chelators for [111In]In3+, suggesting their value for SPECT radiopharmaceuticals.
Collapse
Affiliation(s)
- Kevin K Lee
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA.
| | - Mou Chakraborty
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
- Program in Quantitative Molecular Therapeutics, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - Aohan Hu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA.
| | - Thines Kanagasundaram
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA.
| | - Daniel L J Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
- Program in Quantitative Molecular Therapeutics, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, 63110, USA
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA.
| |
Collapse
|
7
|
Koniar H, Wharton L, Ingham A, Rodríguez-Rodríguez C, Kunz P, Radchenko V, Yang H, Rahmim A, Uribe C, Schaffer P. In vivoquantitative SPECT imaging of actinium-226: feasibility and proof-of-concept. Phys Med Biol 2024; 69:155003. [PMID: 38925140 DOI: 10.1088/1361-6560/ad5c37] [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] [Received: 04/24/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Objective.225Ac radiopharmaceuticals have tremendous potential for targeted alpha therapy, however,225Ac (t1/2= 9.9 d) lacks direct gamma emissions forin vivoimaging.226Ac (t1/2= 29.4 h) is a promising element-equivalent matched diagnostic radionuclide for preclinical evaluation of225Ac radiopharmaceuticals.226Ac has two gamma emissions (158 keV and 230 keV) suitable for SPECT imaging. This work is the first feasibility study forin vivoquantitative226Ac SPECT imaging and validation of activity estimation.Approach.226Ac was produced at TRIUMF (Vancouver, Canada) with its Isotope Separator and Accelerator (ISAC) facility. [226Ac]Ac3+was radiolabelled with the bioconjugate crown-TATE developed for therapeutic targeting of neuroendocrine tumours. Mice with AR42J tumour xenografts were injected with either 2 MBq of [226Ac]Ac-crown-TATE or 4 MBq of free [226Ac]Ac3+activity and were scanned at 1, 2.5, 5, and 24 h post injection in a preclinical microSPECT/CT. Quantitative SPECT images were reconstructed from the 158 keV and 230 keV photopeaks with attenuation, background, and scatter corrections. Image-based226Ac activity measurements were assessed from volumes of interest within tumours and organs of interest. Imaging data was compared withex vivobiodistribution measured via gamma counter.Main results. We present, to the best of our knowledge, the first everin vivoquantitative SPECT images of226Ac activity distributions. Time-activity curves derived from SPECT images quantify thein vivobiodistribution of [226Ac]Ac-crown-TATE and free [226Ac]Ac3+activity. Image-based activity measurements in the tumours and organs of interest corresponded well withex vivobiodistribution measurements.Significance. Here in, we established the feasibility ofin vivo226Ac quantitative SPECT imaging for accurate measurement of actinium biodistribution in a preclinical model. This imaging method could facilitate more efficient development of novel actinium labelled compounds by providing accurate quantitativein vivopharmacokinetic information essential for estimating toxicities, dosimetry, and therapeutic potency.
Collapse
Affiliation(s)
- Helena Koniar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Aidan Ingham
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Cristina Rodríguez-Rodríguez
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Peter Kunz
- Accelerator Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Arman Rahmim
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
- Department of Functional Imaging, BC Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| |
Collapse
|
8
|
Toyohara J, Vugts D, Kiss OC, Todde S, Li XG, Liu Z, Yang Z, Gillings N, Cazzola E, Szymanski W, Meulen NVD, Reilly R, Taddei C, Schirrmacher R, Li Z, Lagebo YJ, Bentaleb N, Souza Albernaz MD, Lapi S, Ramogida C, Mukherjee A, Ajenjo J, Deuther-Conrad W, Bourdeau C. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2024; 9:42. [PMID: 38753262 PMCID: PMC11098975 DOI: 10.1186/s41181-024-00268-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development. MAIN BODY This selection of highlights provides commentary on 24 different topics selected by each coauthoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field in many aspects.
Collapse
Affiliation(s)
- Jun Toyohara
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Danielle Vugts
- Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Oliver C Kiss
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
| | - Sergio Todde
- University of Milano-Bicoccia, Tecnomed Foundation, Monza, Italy
| | - Xiang-Guo Li
- Turku PET Centre and Department of Chemistry, and InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | | | - Zhi Yang
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Nic Gillings
- Copenhagen University Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | - Naoual Bentaleb
- National Center for Nuclear Energy, Science and Technology-CNESTEN, Rabat, Morocco
| | - Marta de Souza Albernaz
- University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suzanne Lapi
- University of Alabama at Birmingham, Birmingham, USA
| | - Caterina Ramogida
- Simon Fraser University, Burnaby, Vancouver, Canada
- TRIUMF, Burnaby, Vancouver, Canada
| | - Archana Mukherjee
- Bhabha Atomic Research Center and Homi Bhabha National Institute, Mumbai, India
| | - Javier Ajenjo
- Molecular Imaging Program at Stanford (MIPS), Dept of Radiology, School of Medicine, Stanford University, Stanford, CA), USA
| | - Winnie Deuther-Conrad
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Leipzig, Germany
| | | |
Collapse
|
9
|
Bauer D, De Gregorio R, Pratt EC, Bell A, Michel A, Lewis JS. Exploring the PET in vivo generator 134Ce as a theranostic match for 225Ac. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591165. [PMID: 38712285 PMCID: PMC11071455 DOI: 10.1101/2024.04.25.591165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Purpose The radionuclide pair cerium-134/lanthanum-134 (134Ce/134La) was recently proposed as a suitable diagnostic counterpart for the therapeutic alpha-emitter actinium-225 (225Ac). The unique properties of 134Ce offer perspectives for developing innovative in vivo investigations not possible with 225Ac. In this work, 225Ac- and 134Ce-labeled tracers were directly compared using internalizing and slow-internalizing cancer models to evaluate their in vivo comparability, progeny meandering, and potential as a matched theranostic pair for clinical translation. Despite being an excellent chemical match, 134Ce/134La has limitations to the setting of quantitative positron emission tomography imaging. Methods The precursor PSMA-617 and a macropa-based tetrazine-conjugate (mcp-PEG8-Tz) were radiolabelled with 225Ac or 134Ce and compared in vitro and in vivo using standard (radio)chemical methods. Employing biodistribution studies and positron emission tomography (PET) imaging in athymic nude mice, the radiolabelled PSMA-617 tracers were evaluated in a PC3/PIP (PC3 engineered to express a high level of prostate-specific membrane antigen) prostate cancer mouse model. The 225Ac and 134Ce-labeled mcp-PEG8-Tz were investigated in a BxPC-3 pancreatic tumour model harnessing the pretargeting strategy based on a trans-cyclooctene-modified 5B1 monoclonal antibody. Results In vitro and in vivo studies with both 225Ac and 134Ce-labelled tracers led to comparable results, confirming the matching pharmacokinetics of this theranostic pair. However, PET imaging of the 134Ce-labelled precursors indicated that quantification is highly dependent on tracer internalization due to the redistribution of 134Ce's PET-compatible daughter 134La. Consequently, radiotracers based on internalizing vectors like PSMA-617 are suited for this theranostic pair, while slow-internalizing 225Ac-labelled tracers are not quantitatively represented by 134Ce PET imaging. Conclusion When employing slow-internalizing vectors, 134Ce might not be an ideal match for 225Ac due to the underestimation of tumour uptake caused by the in vivo redistribution of 134La. However, this same characteristic makes it possible to estimate the redistribution of 225Ac's progeny noninvasively. In future studies, this unique PET in vivo generator will further be harnessed to study tracer internalization, trafficking of receptors, and the progression of the tumour microenvironment.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Roberto De Gregorio
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Edwin C. Pratt
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abram Bell
- Brigham Young University-Idaho, Rexburg, ID 83440, USA
| | - Alexa Michel
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason S. Lewis
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Departments of Radiology and Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
10
|
Wadhwa A, Wang S, Patiño-Escobar B, Bidkar AP, Bobba KN, Chan E, Meher N, Bidlingmaier S, Su Y, Dhrona S, Geng H, Sarin V, VanBrocklin HF, Wilson DM, He J, Zhang L, Steri V, Wong SW, Martin TG, Seo Y, Liu B, Wiita AP, Flavell RR. CD46-Targeted Theranostics for PET and 225Ac-Radiopharmaceutical Therapy of Multiple Myeloma. Clin Cancer Res 2024; 30:1009-1021. [PMID: 38109209 PMCID: PMC10905524 DOI: 10.1158/1078-0432.ccr-23-2130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/26/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE Multiple myeloma is a plasma cell malignancy with an unmet clinical need for improved imaging methods and therapeutics. Recently, we identified CD46 as an overexpressed therapeutic target in multiple myeloma and developed the antibody YS5, which targets a cancer-specific epitope on this protein. We further developed the CD46-targeting PET probe [89Zr]Zr-DFO-YS5 for imaging and [225Ac]Ac-DOTA-YS5 for radiopharmaceutical therapy of prostate cancer. These prior studies suggested the feasibility of the CD46 antigen as a theranostic target in multiple myeloma. Herein, we validate [89Zr]Zr-DFO-YS5 for immunoPET imaging and [225Ac]Ac-DOTA-YS5 for radiopharmaceutical therapy of multiple myeloma in murine models. EXPERIMENTAL DESIGN In vitro saturation binding was performed using the CD46 expressing MM.1S multiple myeloma cell line. ImmunoPET imaging using [89Zr]Zr-DFO-YS5 was performed in immunodeficient (NSG) mice bearing subcutaneous and systemic multiple myeloma xenografts. For radioligand therapy, [225Ac]Ac-DOTA-YS5 was prepared, and both dose escalation and fractionated dose treatment studies were performed in mice bearing MM1.S-Luc systemic xenografts. Tumor burden was analyzed using BLI, and body weight and overall survival were recorded to assess antitumor effect and toxicity. RESULTS [89Zr]Zr-DFO-YS5 demonstrated high affinity for CD46 expressing MM.1S multiple myeloma cells (Kd = 16.3 nmol/L). In vitro assays in multiple myeloma cell lines demonstrated high binding, and bioinformatics analysis of human multiple myeloma samples revealed high CD46 expression. [89Zr]Zr-DFO-YS5 PET/CT specifically detected multiple myeloma lesions in a variety of models, with low uptake in controls, including CD46 knockout (KO) mice or multiple myeloma mice using a nontargeted antibody. In the MM.1S systemic model, localization of uptake on PET imaging correlated well with the luciferase expression from tumor cells. A treatment study using [225Ac]Ac-DOTA-YS5 in the MM.1S systemic model demonstrated a clear tumor volume and survival benefit in the treated groups. CONCLUSIONS Our study showed that the CD46-targeted probe [89Zr]Zr-DFO-YS5 can successfully image CD46-expressing multiple myeloma xenografts in murine models, and [225Ac]Ac-DOTA-YS5 can effectively inhibit the growth of multiple myeloma. These results demonstrate that CD46 is a promising theranostic target for multiple myeloma, with the potential for clinical translation.
Collapse
Affiliation(s)
- Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Bonell Patiño-Escobar
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Emily Chan
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, California
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Vishesh Sarin
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | - Li Zhang
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Medicine, Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Veronica Steri
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Sandy W. Wong
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Thomas G. Martin
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Bin Liu
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Anesthesia, University of California, San Francisco, California
| | - Arun P. Wiita
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
- Chan Zuckerberg Biohub, San Francisco, California
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California
| |
Collapse
|
11
|
Bobba KN, Bidkar AP, Wadhwa A, Meher N, Drona S, Sorlin AM, Bidlingmaier S, Zhang L, Wilson DM, Chan E, Greenland NY, Aggarwal R, VanBrocklin HF, He J, Chou J, Seo Y, Liu B, Flavell RR. Development of CD46 targeted alpha theranostics in prostate cancer using 134Ce/ 225Ac-Macropa-PEG 4-YS5. Theranostics 2024; 14:1344-1360. [PMID: 38389832 PMCID: PMC10879874 DOI: 10.7150/thno.92742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: 225Ac, a long-lived α-emitter with a half-life of 9.92 days, has garnered significant attention as a therapeutic radionuclide when coupled with monoclonal antibodies and other targeting vectors. Nevertheless, its clinical utility has been hampered by potential off-target toxicity, a lack of optimized chelators for 225Ac, and limitations in radiolabeling methods. In a prior study evaluating the effectiveness of CD46-targeted radioimmunotherapy, we found great therapeutic efficacy but also significant toxicity at higher doses. To address these challenges, we have developed a radioimmunoconjugate called 225Ac-Macropa-PEG4-YS5, incorporating a stable PEGylated linker to maximize tumoral uptake and increase tumor-to-background ratios. Our research demonstrates that this conjugate exhibits greater anti-tumor efficacy while minimizing toxicity in prostate cancer 22Rv1 tumors. Methods: We synthesized Macropa.NCS and Macropa-PEG4/8-TFP esters and prepared Macropa-PEG0/4/8-YS5 (with nearly ~1:1 ratio of macropa chelator to antibody YS5) as well as DOTA-YS5 conjugates. These conjugates were then radiolabeled with 225Ac in a 2 M NH4OAc solution at 30 °C, followed by purification using YM30K centrifugal purification. Subsequently, we conducted biodistribution studies and evaluated antitumor activity in nude mice (nu/nu) bearing prostate 22Rv1 xenografts in both single-dose and fractionated dosing studies. Micro-PET imaging studies were performed with 134Ce-Macropa-PEG0/4/8-YS5 in 22Rv1 xenografts for 7 days. Toxicity studies were also performed in healthy athymic nude mice. Results: As expected, we achieved a >95% radiochemical yield when labeling Macropa-PEG0/4/8-YS5 with 225Ac, regardless of the chelator ratios (ranging from 1 to 7.76 per YS5 antibody). The isolated yield exceeded 60% after purification. Such high conversions were not observed with the DOTA-YS5 conjugate, even at a higher ratio of 8.5 chelators per antibody (RCY of 83%, an isolated yield of 40%). Biodistribution analysis at 7 days post-injection revealed higher tumor uptake for the 225Ac-Macropa-PEG4-YS5 (82.82 ± 38.27 %ID/g) compared to other conjugates, namely 225Ac-Macropa-PEG0/8-YS5 (38.2 ± 14.4/36.39 ± 12.4 %ID/g) and 225Ac-DOTA-YS5 (29.35 ± 7.76 %ID/g). The PET Imaging of 134Ce-Macropa-PEG0/4/8-YS5 conjugates resulted in a high tumor uptake, and tumor to background ratios. In terms of antitumor activity, 225Ac-Macropa-PEG4-YS5 exhibited a substantial response, leading to prolonged survival compared to 225Ac-DOTA-YS5, particularly when administered at 4.625 kBq doses, in single or fractionated dose regimens. Chronic toxicity studies observed mild to moderate renal toxicity at 4.625 and 9.25 kBq doses. Conclusions: Our study highlights the promise of 225Ac-Macropa-PEG4-YS5 for targeted alpha particle therapy. The 225Ac-Macropa-PEG4-YS5 conjugate demonstrates improved biodistribution, reduced off-target binding, and enhanced therapeutic efficacy, particularly at lower doses, compared to 225Ac-DOTA-YS5. Incorporating theranostic 134Ce PET imaging further enhances the versatility of macropa-PEG conjugates, offering a more effective and safer approach to cancer treatment. Overall, this methodology has a high potential for broader clinical applications.
Collapse
Affiliation(s)
- Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Suchi Drona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Alexandre M. Sorlin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, California 94110, United States
| | - Li Zhang
- Department of Medicine and the Department of Epidemiology and Biostatistics, University of California, Berkeley, California, United States
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
| | - Emily Chan
- Department of Pathology, University of California, San Francisco, California 94110, United States
| | - Nancy Y. Greenland
- Department of Pathology, University of California, San Francisco, California 94110, United States
| | - Rahul Aggarwal
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, United States
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, 22908, United States
| | - Jonathan Chou
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, California, United States
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, California 94110, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California 94143, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California 94143-0981, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158-2517, United States
| |
Collapse
|
12
|
Jalloul W, Ghizdovat V, Stolniceanu CR, Ionescu T, Grierosu IC, Pavaleanu I, Moscalu M, Stefanescu C. Targeted Alpha Therapy: All We Need to Know about 225Ac's Physical Characteristics and Production as a Potential Theranostic Radionuclide. Pharmaceuticals (Basel) 2023; 16:1679. [PMID: 38139806 PMCID: PMC10747780 DOI: 10.3390/ph16121679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The high energy of α emitters, and the strong linear energy transfer that goes along with it, lead to very efficient cell killing through DNA damage. Moreover, the degree of oxygenation and the cell cycle state have no impact on these effects. Therefore, α radioisotopes can offer a treatment choice to individuals who are not responding to β- or gamma-radiation therapy or chemotherapy drugs. Only a few α-particle emitters are suitable for targeted alpha therapy (TAT) and clinical applications. The majority of available clinical research involves 225Ac and its daughter nuclide 213Bi. Additionally, the 225Ac disintegration cascade generates γ decays that can be used in single-photon emission computed tomography (SPECT) imaging, expanding the potential theranostic applications in nuclear medicine. Despite the growing interest in applying 225Ac, the restricted global accessibility of this radioisotope makes it difficult to conduct extensive clinical trials for many radiopharmaceutical candidates. To boost the availability of 225Ac, along with its clinical and potential theranostic applications, this review attempts to highlight the fundamental physical properties of this α-particle-emitting isotope, as well as its existing and possible production methods.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Teodor Ionescu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana Pavaleanu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| |
Collapse
|
13
|
Pretze M, Michler E, Runge R, Wetzig K, Tietze K, Brandt F, Schultz MK, Kotzerke J. Influence of the Molar Activity of 203/212Pb-PSC-PEG 2-TOC on Somatostatin Receptor Type 2-Binding and Cell Uptake. Pharmaceuticals (Basel) 2023; 16:1605. [PMID: 38004470 PMCID: PMC10675797 DOI: 10.3390/ph16111605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: In neuroendocrine tumors (NETs), somatostatin receptor subtype 2 is highly expressed, which can be targeted by a radioactive ligand such as [177Lu]Lu-1,4,7,10-tetraazacyclododecane-N,N',N″,N‴,-tetraacetic acid-[Tyr3,Thr8]-octreotide (177Lu-DOTA-TOC) and, more recently, by a lead specific chelator (PSC) containing 203/212Pb-PSC-PEG2-TOC (PSC-TOC). The molar activity (AM) can play a crucial role in tumor uptake, especially in receptor-mediated uptake, such as in NETs. Therefore, an investigation of the influence of different molar activities of 203/212Pb-PSC-TOC on cell uptake was investigated. (2) Methods: Optimized radiolabeling of 203/212Pb-PSC-TOC was performed with 50 µg of precursor in a NaAc/AcOH buffer at pH 5.3-5.5 within 15-45 min at 95° C. Cell uptake was studied in AR42 J, HEK293 sst2, and ZR75-1 cells. (3) Results: 203/212Pb-PSC-TOC was radiolabeled with high radiochemical purity >95% and high radiochemical yield >95%, with AM ranging from 0.2 to 61.6 MBq/nmol. The cell uptake of 203Pb-PSC-TOC (AM = 38 MBq/nmol) was highest in AR42 J (17.9%), moderate in HEK293 sstr (9.1%) and lowest in ZR75-1 (0.6%). Cell uptake increased with the level of AM. (4) Conclusions: A moderate AM of 15-40 MBq/nmol showed the highest cell uptake. No uptake limitation was found in the first 24-48 h. Further escalation experiments with even higher AM should be performed in the future. It was shown that AM plays an important role because of its direct dependence on the cellular uptake levels, possibly due to less receptor saturation with non-radioactive ligands at higher AM.
Collapse
Affiliation(s)
- Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Enrico Michler
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Roswitha Runge
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Kerstin Wetzig
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Katja Tietze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Florian Brandt
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Michael K. Schultz
- Department of Radiology, University of Iowa, Iowa City, IA 52240, USA;
- Viewpoint Molecular Targeting, Inc. (DBA Perspective Therapeutics), Coralville, IA 52241, USA
- Department of Chemistry, University of Iowa, Iowa City, IA 52241, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| |
Collapse
|
14
|
Jang A, Kendi AT, Johnson GB, Halfdanarson TR, Sartor O. Targeted Alpha-Particle Therapy: A Review of Current Trials. Int J Mol Sci 2023; 24:11626. [PMID: 37511386 PMCID: PMC10380274 DOI: 10.3390/ijms241411626] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Radiopharmaceuticals are rapidly developing as a field, with the successful use of targeted beta emitters in neuroendocrine tumors and prostate cancer serving as catalysts. Targeted alpha emitters are in current development for several potential oncologic indications. Herein, we review the three most prevalently studied conjugated/chelated alpha emitters (225actinium, 212lead, and 211astatine) and focus on contemporary clinical trials in an effort to more fully appreciate the breadth of the current evaluation. Phase I trials targeting multiple diseases are now underway, and at least one phase III trial (in selected neuroendocrine cancers) is currently in the initial stages of recruitment. Combination trials are now also emerging as alpha emitters are integrated with other therapies in an effort to create solutions for those with advanced cancers. Despite the promise of targeted alpha therapies, many challenges remain. These challenges include the development of reliable supply chains, the need for a better understanding of the relationships between administered dose and absorbed dose in both tissue and tumor and how that predicts outcomes, and the incomplete understanding of potential long-term deleterious effects of the alpha emitters. Progress on multiple fronts is necessary to bring the potential of targeted alpha therapies into the clinic.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ayse T Kendi
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Oliver Sartor
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
15
|
Klauser PC, Chopra S, Cao L, Bobba KN, Yu B, Seo Y, Chan E, Flavell RR, Evans MJ, Wang L. Covalent Proteins as Targeted Radionuclide Therapies Enhance Antitumor Effects. ACS CENTRAL SCIENCE 2023; 9:1241-1251. [PMID: 37396859 PMCID: PMC10311652 DOI: 10.1021/acscentsci.3c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Indexed: 07/04/2023]
Abstract
Molecularly targeted radionuclide therapies (TRTs) struggle with balancing efficacy and safety, as current strategies to increase tumor absorption often alter drug pharmacokinetics to prolong circulation and normal tissue irradiation. Here we report the first covalent protein TRT, which, through reacting with the target irreversibly, increases radioactive dose to the tumor without altering the drug's pharmacokinetic profile or normal tissue biodistribution. Through genetic code expansion, we engineered a latent bioreactive amino acid into a nanobody, which binds to its target protein and forms a covalent linkage via the proximity-enabled reactivity, cross-linking the target irreversibly in vitro, on cancer cells, and on tumors in vivo. The radiolabeled covalent nanobody markedly increases radioisotope levels in tumors and extends tumor residence time while maintaining rapid systemic clearance. Furthermore, the covalent nanobody conjugated to the α-emitter actinium-225 inhibits tumor growth more effectively than the noncovalent nanobody without causing tissue toxicity. Shifting the protein-based TRT from noncovalent to covalent mode, this chemical strategy improves tumor responses to TRTs and can be readily scaled to diverse protein radiopharmaceuticals engaging broad tumor targets.
Collapse
Affiliation(s)
- Paul C. Klauser
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Shalini Chopra
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Li Cao
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Kondapa Naidu Bobba
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Bingchen Yu
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| | - Youngho Seo
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Emily Chan
- Department
of Pathology, University of California San
Francisco, San Francisco, California 94158, United States
| | - Robert R. Flavell
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Michael J. Evans
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
- Department
of Radiology and Biomedical Imaging, University
of California San Francisco, San Francisco, California 94158, United States
| | - Lei Wang
- Department
of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, California 94158, United States
- Helen
Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|