1
|
Bogdanovic B, Hugonnet F, Montemagno C. Theranostics in Hematological Malignancies: Cutting-Edge Advances in Diagnosis and Targeted Therapy. Cancers (Basel) 2025; 17:1247. [PMID: 40227793 PMCID: PMC11987953 DOI: 10.3390/cancers17071247] [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: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025] Open
Abstract
Hematologic malignancies, including leukemia, lymphoma, and multiple myeloma, continue to challenge clinicians with complex treatment regimens that often involve significant side effects and limited success, especially in advanced stages. Recent advancements in nuclear medicine have introduced theranostic strategies that merge diagnostic imaging with targeted therapeutic approaches, offering the potential for more precise and personalized treatment. A key area of progress lies in the development of alpha-emitting radiopharmaceuticals, such as 225Ac, 211At, or 212Pb, which can deliver potent radiation directly to tumor cells, sparing healthy tissue and minimizing collateral damage. In parallel with these therapeutic advancements, molecular imaging using radiolabeled agents enables better disease monitoring, assessment of treatment efficacy, and personalized management of patients with hematologic malignancies. The integration of diagnostic imaging with radiotherapy allows for a more tailored approach, where treatment can be adjusted based on real-time information about tumor progression and response. This review examines the recent strides made in both the development of radiopharmaceuticals and their applications in molecular imaging, with a focus on the potential to improve precision, reduce toxicity, and optimize patient outcomes. The synergy between targeted therapy and molecular imaging represents a transformative shift in the management of hematologic malignancies. As these technologies evolve, they are poised to redefine treatment paradigms, offering new hope for patients and potentially improving survival rates with more effective and less toxic treatment options.
Collapse
Affiliation(s)
- Bojana Bogdanovic
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Florent Hugonnet
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Christopher Montemagno
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|
2
|
Plichta KA, Buatti JM. The Emerging Potential of Lead-212 Theranostics. Hematol Oncol Clin North Am 2025; 39:221-236. [PMID: 39827042 DOI: 10.1016/j.hoc.2024.11.001] [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: 01/22/2025]
Abstract
The field of theranostics uses radiopharmaceuticals to diagnose and treat disease, allowing for a personalized approach to treatment. Most theranostic therapies involve the use of beta-emitting radiopharmaceuticals. Because of their higher energies and decreased range, the use of alpha-emitting radiopharmaceuticals offers potential advantages over beta-emitting radiopharmaceuticals, including the potential for improved cell kill and decreased toxicity to normal tissues. This article focuses on the potential use of lead-212 as a theranostic treatment agent.
Collapse
Affiliation(s)
- Kristin A Plichta
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
3
|
Goto H, Shiraishi Y, Okada S. Continuing progress in radioimmunotherapy for hematologic malignancies. Blood Rev 2025; 69:101250. [PMID: 39609167 DOI: 10.1016/j.blre.2024.101250] [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: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Abstract
Radioimmunotherapy (RIT) involves combining a cytotoxic radionuclide with an antibody (Ab) targeting a tumor antigen. Compared with conventional therapies, RIT improves the therapeutic efficacy of Ab and ameliorates toxicity. This comprehensive review describes the current advancements and future prospects in RIT for treating hematologic malignancies based on recent investigations. Although β-particle RITs targeting CD20 are effective with low toxicity in patients with relapsed/refractory indolent or transformed non-Hodgkin's lymphoma, these treatments have not gained popularity because of the increasing availability of new therapies. RIT using single-domain antibodies is expected to improve tumor penetrance and reduce radiation exposure to non-target organs. To enhance RIT efficacy, α-particle RIT and pretargeted radioimmunotherapy (PRIT) are currently being developed. Alpha-particle RIT demonstrates substantial antitumor activity and reduced bystander effects due to its high linear energy transfer and short particle range. PRIT may increase the tumor-to-whole body dose ratio.
Collapse
Affiliation(s)
- Hiroki Goto
- Division of Radioisotope and Tumor Pathobiology, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Yoshioki Shiraishi
- Radioisotope Center, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
4
|
Zubenko AD, Pashanova AV, Mosaleva SP, Chernikova EY, Karnoukhova VA, Fedyanin IV, Egorova BV, Shchukina AA, Fedorov YV, Fedorova OA. Double-Armed 18- and 21-Membered Macrocycles as Potential Chelators for Lead and Bismuth Radiopharmaceuticals. Inorg Chem 2024; 63:21652-21669. [PMID: 39475211 DOI: 10.1021/acs.inorgchem.4c03116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
With increasing clinical applications and interest in targeted alpha therapy, there is growing interest in developing alternative chelating agents for [212Pb]Pb2+ and [212/213Bi]Bi3+ that exhibit rapid radiolabeling kinetics and kinetic inertness. Herein we report the synthesis and detailed investigation of diacetate and dipicolinate 18- and 21-membered macrocyclic chelators BADA-18, BADA-21, BADPA-18, and BADPA-21 for the complexation of Pb2+ and Bi3+ ions with potential use in the preparation of radiopharmaceuticals. The formation of mononuclear complexes was established by using ESI-mass spectrometry, and their stability constants were determined by potentiometric titration. A thorough study of the structure of the metal complexes was carried out by using X-ray diffraction and NMR spectroscopy. It was shown how the stability of the complex is influenced by an increase in the size of the macrocycle, the replacement of acetate arms with picolinate ones, the rigidity of the ligand, as well as the type of conformation (syn- or anti-) of the metal complex. The new ligands were radiolabeled with [210Pb]Pb2+ and [207Bi]Bi3+, and the in vitro stability of the resulting complexes in a competitive environment of serum and biologically significant metal ions was assessed. Rapid complex formation in 1-2 min at room temperature, as well as the high kinetic inertness of the complexes Pb(BADPA-18) and Bi(BADPA-18) in biological media, demonstrate its potential for use in targeted radionuclide therapy.
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 119991, Russian Federation
| | - Anna V Pashanova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Sofia P Mosaleva
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
- D. I. Mendeleev University of Chemistry and Technology of Russia, 125047 Miusskaya sqr., 9, Moscow 119991, Russian Federation
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Valentina A Karnoukhova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Ivan V Fedyanin
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Bayirta V Egorova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow 119991, Russian Federation
| | - Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
- D. I. Mendeleev University of Chemistry and Technology of Russia, 125047 Miusskaya sqr., 9, Moscow 119991, Russian Federation
| |
Collapse
|
5
|
Saidi A, Stallons TA, Wong AG, Torgue JJ. Preclinical Investigation of [ 212Pb]Pb-DOTAM-GRPR1 for Peptide Receptor Radionuclide Therapy in a Prostate Tumor Model. J Nucl Med 2024; 65:1769-1775. [PMID: 39327021 PMCID: PMC11533912 DOI: 10.2967/jnumed.124.268101] [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: 05/30/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The role of gastrin-releasing peptide receptor (GRPR) in various diseases, including cancer, has been extensively studied and has emerged as a promising therapeutic target. In this study, we successfully achieved the use of [212Pb]Pb-DOTAM-GRPR1, comprising the α-particle generator, 212Pb, combined with a GRPR-targeting peptide, GRPR1, in a prostate cancer model. Methods: Pharmacokinetics, toxicity, radiation dosimetry, and efficacy were assessed in GRPR-positive prostate tumor-bearing mice after intravenous administration of [212Pb]Pb-DOTAM-GRPR1 (where DOTAM is 1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane). Results: Preclinical studies have shown tumor targeting of up to 5 percent injected dose per gram over 24 h, and optimization of the drug formulation and quantity has led to minimized oxidation and off-target binding, respectively. Particularly, an increase in peptide amount from 28 to 280 ng was shown to reduce off-target uptake, especially at the level of the pancreas, by about 30%. Furthermore, dosimetry studies confirmed the kidney as the dose-limiting organ, and toxicity studies revealed that a nontoxic dose of up to 1,665 kBq could be injected into mice. Efficacy studies indicated a median survival time of 9 wk in the control group, which received only a buffer solution, compared with 19 wk in the group that received 4 injections of 370 kBq at 3-wk intervals. Conclusion: Taken together, these combined data demonstrate the safety, tolerability, and efficacy of [212Pb]Pb-DOTAM-GRPR1, thus warranting further exploration in clinical trials.
Collapse
|
6
|
Lindland K, Malenge MM, Li RG, Wouters R, Bønsdorff TB, Juzeniene A, Dragovic SM. Antigen targeting and anti-tumor activity of a novel anti-CD146 212Pb internalizing alpha-radioimmunoconjugate against malignant peritoneal mesothelioma. Sci Rep 2024; 14:25941. [PMID: 39472474 PMCID: PMC11522520 DOI: 10.1038/s41598-024-76778-z] [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: 06/20/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Malignant mesothelioma, a highly aggressive cancer that primarily affects the serosal membranes, has limited therapeutic options, particularly for cavitary tumors, such as peritoneal and pleural malignant mesothelioma. Intracavitary administration of a radioimmunoconjugate to locally target mesothelioma cancer cells has been proposed as a treatment. CD146, upregulated in mesothelioma but not in healthy tissues, is a promising therapeutic target. This study characterized CD146 expression and binding/internalization kinetics of the CD146-targeting antibody OI-3 coupled with 212Pb (212Pb-TCMC-OI-3) in human mesothelioma cells. Flow cytometry showed that both chimeric (chOI-3) and murine (mOI-3) antibodies rapidly bound and internalized within 1-6 h in MSTO-211H cells. 212Pb-TCMC-chOI-3 exhibited 3.1- to 13.7-fold and 3.1- to 8.5-fold increased internalized 212Pb and 212Bi atoms per cell at 2 and 24 h, respectively, compared to isotype control, underscoring enhanced internalization efficiency. Intraperitoneal administration of 212Pb-TCMC-mOI-3 to mice with intraperitoneal MSTO-211H xenografts improved median survival by a ratio of 1.3 compared to non-binding 212Pb-TCMC-mIgG1. The ability of 212Pb-TCMC-mOI-3 to target and inhibit the growth of intraperitoneal mesothelioma xenografts supports targeted radionuclide therapy's efficacy for metastatic peritoneal mesothelioma. This study highlights the potential of localized CD146-targeted radioimmunotherapy for malignant mesothelioma, offering a new avenue for improving patient outcomes.
Collapse
Affiliation(s)
- Kim Lindland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0316, Oslo, Norway.
- Department of Radiation Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway.
- Oncoinvent ASA, 0484, Oslo, Norway.
| | | | | | - Roxanne Wouters
- Oncoinvent ASA, 0484, Oslo, Norway
- Laboratory of Tumour Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000, Leuven, Belgium
| | | | - Asta Juzeniene
- Department of Radiation Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | | |
Collapse
|
7
|
Gonzalez-Kozlova E, Huang HH, Jagede OA, Tuballes K, Del Valle DM, Kelly G, Patel M, Xie H, Harris J, Argueta K, Nie K, Barcessat V, Moravec R, Altreuter J, Duose DY, Kahl BS, Ansell SM, Yu J, Cerami E, Lindsay JR, Wistuba II, Kim-Schulze S, Diefenbach CS, Gnjatic S. Tumor-Immune Signatures of Treatment Resistance to Brentuximab Vedotin with Ipilimumab and/or Nivolumab in Hodgkin Lymphoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1726-1737. [PMID: 38934093 PMCID: PMC11247952 DOI: 10.1158/2767-9764.crc-24-0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
To investigate the cellular and molecular mechanisms associated with targeting CD30-expressing Hodgkin lymphoma (HL) and immune checkpoint modulation induced by combination therapies of CTLA4 and PD1, we leveraged Phase 1/2 multicenter open-label trial NCT01896999 that enrolled patients with refractory or relapsed HL (R/R HL). Using peripheral blood, we assessed soluble proteins, cell composition, T-cell clonality, and tumor antigen-specific antibodies in 54 patients enrolled in the phase 1 component of the trial. NCT01896999 reported high (>75%) overall objective response rates with brentuximab vedotin (BV) in combination with ipilimumab (I) and/or nivolumab (N) in patients with R/R HL. We observed a durable increase in soluble PD1 and plasmacytoid dendritic cells as well as decreases in plasma CCL17, ANGPT2, MMP12, IL13, and CXCL13 in N-containing regimens (BV + N and BV + I + N) compared with BV + I (P < 0.05). Nonresponders and patients with short progression-free survival showed elevated CXCL9, CXCL13, CD5, CCL17, adenosine-deaminase, and MUC16 at baseline or after one treatment cycle and a higher prevalence of NY-ESO-1-specific autoantibodies (P < 0.05). The results suggest a circulating tumor-immune-derived signature of BV ± I ± N treatment resistance that may be useful for patient stratification in combination checkpoint therapy. SIGNIFICANCE Identification of multi-omic immune markers from peripheral blood may help elucidate resistance mechanisms to checkpoint inhibitor and antibody-drug conjugate combinations with potential implications for treatment decisions in relapsed HL.
Collapse
Affiliation(s)
- Edgar Gonzalez-Kozlova
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Hsin-Hui Huang
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Opeyemi A. Jagede
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Kevin Tuballes
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Diane M. Del Valle
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Geoffrey Kelly
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Hui Xie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Jocelyn Harris
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Kimberly Argueta
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Vanessa Barcessat
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Radim Moravec
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, Bethesda, Maryland.
| | - Jennifer Altreuter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Dzifa Y. Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Brad S. Kahl
- Washington University School of Medicine, New York, New York.
| | | | - Joyce Yu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - James R. Lindsay
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
- CIMAC-CIDC Network, Pipeline Development and Portal Integration, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Ignacio I. Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Seunghee Kim-Schulze
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| | | | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
8
|
Goto H, Shiraishi Y, Okada S. Recent preclinical and clinical advances in radioimmunotherapy for non-Hodgkin's lymphoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:208-224. [PMID: 38464386 PMCID: PMC10918239 DOI: 10.37349/etat.2024.00213] [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: 10/14/2023] [Accepted: 12/28/2023] [Indexed: 03/12/2024] Open
Abstract
Radioimmunotherapy (RIT) is a therapy that combines a radioactive nucleotide with a monoclonal antibody (mAb). RIT enhances the therapeutic effect of mAb and reduces toxicity compared with conventional treatment. The purpose of this review is to summarize the current progress of RIT for treating non-Hodgkin's lymphoma (NHL) based on recent preclinical and clinical studies. The efficacy of RIT targeting the B-lymphocyte antigen cluster of differentiation 20 (CD20) has been demonstrated in clinical trials. Two radioimmunoconjugates targeting CD20, yttrium-90 (90Y)-ibritumomab-tiuxetan (Zevalin) and iodine-131 (131I)-tositumomab (Bexxar), have been approved in the USA Food and Drug Administration (FDA) for treating relapsed/refractory indolent or transformed NHL in 2002 and 2003, respectively. Although these two radioimmunoconjugates are effective and least toxic, they have not achieved popularity due to increasing access to novel therapies and the complexity of their delivery process. RIT is constantly evolving with the identification of novel targets and novel therapeutic strategies using newer radionuclides such as alpha-particle isotopes. Alpha-particles show very short path lengths and high linear energy transfer. These characteristics provide increased tumor cell-killing activities and reduced non-specific bystander responses on normal tissue. This review also discusses reviewed pre-targeted RIT (PRIT) and immuno-positron emission tomography (PET). PRIT potentially increases the dose of radionuclide delivered to tumors while toxicities to normal tissues are limited. Immuno-PET is a molecular imaging tracer that combines the high sensitivity of PET with the specific targeting capability of mAb. Immuno-PET strategies targeting CD20 and other antigens are currently being developed. The theragnostic approach by immuno-PET will be useful in monitoring the treatment response.
Collapse
Affiliation(s)
- Hiroki Goto
- Division of Radioisotope and Tumor Pathobiology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yoshioki Shiraishi
- Radioisotope Center, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
9
|
Cicone F, Santo G, Bodet-Milin C, Cascini GL, Kraeber-Bodéré F, Stokke C, Kolstad A. Radioimmunotherapy of Non-Hodgkin B-cell Lymphoma: An update. Semin Nucl Med 2023; 53:413-425. [PMID: 36635112 DOI: 10.1053/j.semnuclmed.2022.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023]
Abstract
Systemic radioimmunotherapy (RIT) is arguably the most effective and least toxic anticancer treatment for non-Hodgkin lymphoma (NHL). In treatment-naïve patients with indolent NHL, the efficacy of a single injection of RIT compares with that of multiple cycles of combination chemotherapy. However, 20 years following the approval of the first CD20-targeting radioimmunoconjugates 90Y-Ibritumomab-tiuxetan (Zevalin) and 131I-tositumomab (Bexxar), the number of patients referred for RIT in western countries has dramatically decreased. Notwithstanding this, the development of RIT has continued. Therapeutic targets other than CD20 have been identified, new vector molecules have been produced allowing for faster delivery of RIT to the target, and innovative radionuclides with favorable physical characteristics such as alpha emitters have been more widely available. In this article, we reviewed the current status of RIT in NHL, with particular focus on recent clinical and preclinical developments.
Collapse
Affiliation(s)
- Francesco Cicone
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy; Nuclear Medicine Unit, University Hospital "Mater Domini", Catanzaro, Italy.
| | - Giulia Santo
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Caroline Bodet-Milin
- Nuclear Medicine Department, Nantes Université, Univ Angers, CHU Nantes, INSERM, CNRS, CRCI2NA, F-44000 Nantes, France
| | - Giuseppe Lucio Cascini
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy; Nuclear Medicine Unit, University Hospital "Mater Domini", Catanzaro, Italy
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine Department, Nantes Université, Univ Angers, CHU Nantes, INSERM, CNRS, CRCI2NA, F-44000 Nantes, France
| | - Caroline Stokke
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway; Department of Physics, University of Oslo, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Innlandet Hospital Trust Division Gjøvik, Lillehammer, Norway
| |
Collapse
|
10
|
Li J, Huang T, Hua J, Wang Q, Su Y, Chen P, Bidlingmaier S, Li A, Xie Z, Bidkar AP, Shen S, Shi W, Seo Y, Flavell RR, Gioeli D, Dreicer R, Li H, Liu B, He J. CD46 targeted 212Pb alpha particle radioimmunotherapy for prostate cancer treatment. J Exp Clin Cancer Res 2023; 42:61. [PMID: 36906664 PMCID: PMC10007843 DOI: 10.1186/s13046-023-02636-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/01/2023] [Indexed: 03/13/2023] Open
Abstract
We recently identified CD46 as a novel prostate cancer cell surface antigen that shows lineage independent expression in both adenocarcinoma and small cell neuroendocrine subtypes of metastatic castration resistant prostate cancer (mCRPC), discovered an internalizing human monoclonal antibody YS5 that binds to a tumor selective CD46 epitope, and developed a microtubule inhibitor-based antibody drug conjugate that is in a multi-center phase I trial for mCRPC (NCT03575819). Here we report the development of a novel CD46-targeted alpha therapy based on YS5. We conjugated 212Pb, an in vivo generator of alpha-emitting 212Bi and 212Po, to YS5 through the chelator TCMC to create the radioimmunoconjugate, 212Pb-TCMC-YS5. We characterized 212Pb-TCMC-YS5 in vitro and established a safe dose in vivo. We next studied therapeutic efficacy of a single dose of 212Pb-TCMC-YS5 using three prostate cancer small animal models: a subcutaneous mCRPC cell line-derived xenograft (CDX) model (subcu-CDX), an orthotopically grafted mCRPC CDX model (ortho-CDX), and a prostate cancer patient-derived xenograft model (PDX). In all three models, a single dose of 0.74 MBq (20 µCi) 212Pb-TCMC-YS5 was well tolerated and caused potent and sustained inhibition of established tumors, with significant increases of survival in treated animals. A lower dose (0.37 MBq or 10 µCi 212Pb-TCMC-YS5) was also studied on the PDX model, which also showed a significant effect on tumor growth inhibition and prolongation of animal survival. These results demonstrate that 212Pb-TCMC-YS5 has an excellent therapeutic window in preclinical models including PDXs, opening a direct path for clinical translation of this novel CD46-targeted alpha radioimmunotherapy for mCRPC treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Tao Huang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Chongqing Cancer Hospital, Chongqing University, Chongqing, China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Ping Chen
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Guangdong, 518036, China
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Allan Li
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA
| | - Zhongqiu Xie
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama, Birmingham, AL, 35233, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94110, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA
| | - Daniel Gioeli
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Robert Dreicer
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
- Department of Medicine, University of Virginia, Charlottesville, VA, 22903, USA
| | - Hui Li
- Department of Pathology, University of Virginia, Charlottesville, VA, 22903, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, CA, 94110, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94110, USA.
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, 22903, USA.
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
11
|
Liatsou I, Josefsson A, Yu J, Cortez A, Bastiaannet R, Velarde E, Davis K, Brayton C, Wang H, Torgue J, Hobbs RF, Sgouros G. Bone Marrow Relative Biological Effectiveness for a 212Pb-labeled Anti-HER2/neu Antibody. Int J Radiat Oncol Biol Phys 2023; 115:518-528. [PMID: 35926719 DOI: 10.1016/j.ijrobp.2022.07.1842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE We have determined the in vivo relative biological effectiveness (RBE) of an alpha-particle-emitting radiopharmaceutical therapeutic agent (212Pb-labeled anti-HER2/neu antibody) for the bone marrow, a potentially dose-limiting normal tissue. METHODS AND MATERIALS The RBE was measured in mice using femur marrow cellularity as the biological endpoint. External beam radiation therapy (EBRT), delivered by a small-animal radiation research platform was used as the reference radiation. Alpha-particle emissions were delivered by 212Bi after the decay of its parent nuclide 212Pb, which was conjugated onto an anti-HER2/neu antibody. The alpha-particle absorbed dose to the marrow after an intravenous administration (tail vein) of 122.1 to 921.3 kBq 212Pb-TCMC-7.16.4 was calculated. The mice were sacrificed at 0 to 7 days after treatment and the radioactivity from the femur bone marrow was measured. Changes in marrow cellularity were assessed by histopathology. RESULTS The dose response for EBRT and 212Pb-anti-HER2/neu antibody were linear-quadratic and linear, respectively. On transforming the EBRT dose-response relationship into a linear relationship using the equivalent dose in 2-Gy fractions of external beam radiation formalism, we obtained an RBE (denoted RBE2) of 6.4, which is independent of cellularity and absorbed dose. CONCLUSIONS Because hematologic toxicity is dose limiting in almost all antibody-based RPT, in vivo measurements of RBE are important in helping identify an initial administered activity in phase 1 escalation trials. Applying the RBE2 and assuming typical antibody clearance kinetics (biological half-life of 48 hours), using a modified blood-based dosimetry method, an average administered activity of approximately 185.5 MBq (5.0 mCi) per patient could be administered before hematologic toxicity is anticipated.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Anders Josefsson
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jing Yu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Angel Cortez
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Remco Bastiaannet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kaori Davis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Robert F Hobbs
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Li M, Baumhover NJ, Liu D, Cagle BS, Boschetti F, Paulin G, Lee D, Dai Z, Obot ER, Marks BM, Okeil I, Sagastume EA, Gabr M, Pigge FC, Johnson FL, Schultz MK. Preclinical Evaluation of a Lead Specific Chelator (PSC) Conjugated to Radiopeptides for 203Pb and 212Pb-Based Theranostics. Pharmaceutics 2023; 15:414. [PMID: 36839736 PMCID: PMC9966725 DOI: 10.3390/pharmaceutics15020414] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 01/28/2023] Open
Abstract
203Pb and 212Pb have emerged as promising theranostic isotopes for image-guided α-particle radionuclide therapy for cancers. Here, we report a cyclen-based Pb specific chelator (PSC) that is conjugated to tyr3-octreotide via a PEG2 linker (PSC-PEG-T) targeting somatostatin receptor subtype 2 (SSTR2). PSC-PEG-T could be labeled efficiently to purified 212Pb at 25 °C and also to 212Bi at 80 °C. Efficient radiolabeling of mixed 212Pb and 212Bi in PSC-PEG-T was also observed at 80 °C. Post radiolabeling, stable Pb(II) and Bi(III) radiometal complexes in saline were observed after incubating [203Pb]Pb-PSC-PEG-T for 72 h and [212Bi]Bi-PSC-PEG-T for 5 h. Stable [212Pb]Pb-PSC-PEG-T and progeny [212Bi]Bi-PSC-PEG-T were identified after storage in saline for 24 h. In serum, stable radiometal/radiopeptide were observed after incubating [203Pb]Pb-PSC-PEG-T for 55 h and [212Pb]Pb-PSC-PEG-T for 24 h. In vivo biodistribution of [212Pb]Pb-PSC-PEG-T in tumor-free CD-1 Elite mice and athymic mice bearing AR42J xenografts revealed rapid tumor accumulation, excellent tumor retention and fast renal clearance of both 212Pb and 212Bi, with no in vivo redistribution of progeny 212Bi. Single-photon emission computed tomography (SPECT) imaging of [203Pb]Pb-PSC-PEG-T and [212Pb]Pb-PSC-PEG-T in mice also demonstrated comparable accumulation in AR42J xenografts and renal clearance, confirming the theranostic potential of the elementally identical 203Pb/212Pb radionuclide pair.
Collapse
Affiliation(s)
- Mengshi Li
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | | | - Dijie Liu
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Brianna S. Cagle
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | | | | | - Dongyoul Lee
- Department of Physics and Chemistry, Korea Military Academy, Seoul 01805, Republic of Korea
| | - Zhiming Dai
- Department of Chemistry, The University of Iowa, Iowa City, IA 52240, USA
| | - Ephraim R. Obot
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Brenna M. Marks
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Ibrahim Okeil
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Edwin A. Sagastume
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Moustafa Gabr
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | | | - Frances L. Johnson
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
| | - Michael K. Schultz
- Viewpoint Molecular Targeting, Inc., 2500 Crosspark Road, Coralville, IA 52241, USA
- Department of Radiology, The University of Iowa, Iowa City, IA 52246, USA
- Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
13
|
Bergeron DE, Cessna JT, Fitzgerald RP, Laureano-Pérez L, Pibida L, Zimmerman BE. Primary standardization of 212Pb activity by liquid scintillation counting. Appl Radiat Isot 2022; 190:110473. [PMID: 36201936 PMCID: PMC9827475 DOI: 10.1016/j.apradiso.2022.110473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 01/11/2023]
Abstract
An activity standard for 212Pb in equilibrium with its progeny was realized, based on triple-to-double coincidence ratio (TDCR) liquid scintillation (LS) counting. A Monte Carlo-based approach to estimating uncertainties due to nuclear decay data (branching ratios, beta endpoint energies, γ-ray energies, and conversion coefficients for 212Pb and 208Tl) led to combined standard uncertainties ≤ 0.20 %. Confirmatory primary measurements were made by LS efficiency tracing with tritium and 4παβ(LS)-γ(NaI(Tl)) anticoincidence counting. The standard is discussed in relation to current approaches to 212Pb activity calibration. In particular, potential biases encountered when using inappropriate radionuclide calibrator settings are discussed.
Collapse
Affiliation(s)
- Denis E Bergeron
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA.
| | - Jeffrey T Cessna
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Ryan P Fitzgerald
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Lizbeth Laureano-Pérez
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Leticia Pibida
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Brian E Zimmerman
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| |
Collapse
|
14
|
Tornes AJK, Stenberg VY, Larsen RH, Bruland ØS, Revheim ME, Juzeniene A. Targeted alpha therapy with the 224Ra/ 212Pb-TCMC-TP-3 dual alpha solution in a multicellular tumor spheroid model of osteosarcoma. Front Med (Lausanne) 2022; 9:1058863. [PMID: 36507500 PMCID: PMC9727293 DOI: 10.3389/fmed.2022.1058863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma patients with overt metastases at primary diagnosis have a 5-year survival rate of less than 20%. TP-3 is a murine IgG2b monoclonal antibody with high affinity for an epitope residing on the p80 osteosarcoma cell surface membrane antigen. The tumor-associated antigen p80 is overexpressed in osteosarcomas, and has very low normal tissue expression. We propose a novel dual alpha targeting solution containing two radionuclides from the same decay chain, including the bone-seeking 224Ra, and cancer cell-surface seeking 212Pb-TCMC-TP-3 for the treatment of osteoblastic bone cancers, circulating cancer cells and micrometastases. In this in vitro study, the cytotoxic effects of 212Pb-TCMC-TP-3 (single alpha solution) and 224Ra/212Pb-TCMC-TP-3 (dual alpha solution) were investigated in a multicellular spheroid model mimicking micrometastatic disease in osteosarcoma. OHS spheroids with diameters of 253 ± 98 μm treated with 4.5, 2.7, and 3.3 kBq/ml of 212Pb-TCMC-TP-3 for 1, 4, and 24 h, respectively, were disintegrated within 3 weeks. The 212Pb-TCMC-TP-3 induced a 7-fold delay in spheroid doubling time compared to a 28-times higher dose with the non-specific 212Pb-TCMC-rituximab. The 224Ra/212Pb-TCMC-TP-3 completely disintegrated spheroids with diameters of 218-476 μm within 3 and 2 weeks after 4 and 24 h incubation with 5 kBq/ml, respectively. Treatment with 1 kBq/ml of 224Ra/212Pb-TCMC-TP-3 for 24 h caused an 11.4-fold reduction in spheroid viability compared with unconjugated 224Ra/212Pb. The single and dual alpha solutions with TP-3 showed cytotoxicity in spheroids of clinically relevant size, which warrant further testing of the dual alpha solution using in vivo osteosarcoma models.
Collapse
Affiliation(s)
- Anna Julie Kjøl Tornes
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,ArtBio AS, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,*Correspondence: Anna Julie Kjøl Tornes,
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,ArtBio AS, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Øyvind Sverre Bruland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Kokov KV, Egorova BV, German MN, Klabukov ID, Krasheninnikov ME, Larkin-Kondrov AA, Makoveeva KA, Ovchinnikov MV, Sidorova MV, Chuvilin DY. 212Pb: Production Approaches and Targeted Therapy Applications. Pharmaceutics 2022; 14:189. [PMID: 35057083 PMCID: PMC8777968 DOI: 10.3390/pharmaceutics14010189] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 01/18/2023] Open
Abstract
Over the last decade, targeted alpha therapy has demonstrated its high effectiveness in treating various oncological diseases. Lead-212, with a convenient half-life of 10.64 h, and daughter alpha-emitter short-lived 212Bi (T1/2 = 1 h), provides the possibility for the synthesis and purification of complex radiopharmaceuticals with minimum loss of radioactivity during preparation. As a benefit for clinical implementation, it can be milked from a radionuclide generator in different ways. The main approaches applied for these purposes are considered and described in this review, including chromatographic, solution, and other techniques to isolate 212Pb from its parent radionuclide. Furthermore, molecules used for lead's binding and radiochemical features of preparation and stability of compounds labeled with 212Pb are discussed. The results of preclinical studies with an estimation of therapeutic and tolerant doses as well as recently initiated clinical trials of targeted radiopharmaceuticals are presented.
Collapse
Affiliation(s)
- Konstantin V. Kokov
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Bayirta V. Egorova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Marina N. German
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Ilya D. Klabukov
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249036 Obninsk, Russia;
| | - Michael E. Krasheninnikov
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Antonius A. Larkin-Kondrov
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Kseniya A. Makoveeva
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Michael V. Ovchinnikov
- Laboratory of Peptide Synthesis, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (M.V.O.); (M.V.S.)
| | - Maria V. Sidorova
- Laboratory of Peptide Synthesis, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (M.V.O.); (M.V.S.)
| | - Dmitry Y. Chuvilin
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| |
Collapse
|
16
|
Liatsou I, Yu J, Bastiaannet R, Li Z, Hobbs RF, Torgue J, Sgouros G. 212Pb-conjugated anti-rat HER2/ neu antibody against a neu-N derived murine mammary carcinoma cell line: cell kill and RBE in vitro. Int J Radiat Biol 2022; 98:1452-1461. [PMID: 35073214 PMCID: PMC9673603 DOI: 10.1080/09553002.2022.2033341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE In the current work, the RBE of a 212Pb-conjugated anti-HER2/neu antibody construct has been evaluated, in vitro, by colony formation assay. The RBE was estimated by comparing two absorbed dose-survival curves: the first obtained from the conjugated 212Pb experiments (test radiation), the second obtained by parallel experiments of single bolus irradiation of external beam (reference radiation). MATERIALS AND METHODS Mammary carcinoma NT2.5 cells were treated with (0-3.70) kBq/ml of radiolabeled antibody. Nonspecific binding was assessed with addition of excess amount of unlabeled antibody. The colony formation curves were converted from activity concentration to cell nucleus absorbed dose by simulating the decay and transport of all daughter and secondary particles of 212Pb, using the Monte Carlo code GEANT 4. RESULTS The radiolabeled antibody yielded an RBE of 8.3 at 37% survival and a survival independent RBE (i.e. RBE2) of 9.9. Unbound/untargeted 212Pb-labeled antibody, as obtained in blocking experiments yielded minimal alpha-particle radiation to cells. Conclusions: These results further highlight the importance of specific targeting toward achieving tumor cell kill and low toxicity to normal tissue.
Collapse
Affiliation(s)
- Ioanna Liatsou
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - Jing Yu
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - Remco Bastiaannet
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - Zhi Li
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - Robert F. Hobbs
- Department of Radiation Oncology, School of Medicine, Johns Hopkins University, Baltimore, USA
| | | | - George Sgouros
- Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, USA
| |
Collapse
|
17
|
Jadvar H, Colletti PM. Targeted α-therapy in non-prostate malignancies. Eur J Nucl Med Mol Imaging 2021; 49:47-53. [PMID: 33993386 PMCID: PMC11927099 DOI: 10.1007/s00259-021-05405-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/10/2021] [Indexed: 11/27/2022]
Abstract
Progress in unraveling the complex biology of cancer, novel developments in radiochemistry, and availability of relevant α-emitters for targeted therapy have provided innovative approaches to precision cancer management. The approval of 223Ra dichloride for treatment of men with osseous metastatic castrate-resistant prostate cancer unleashed targeted α-therapy as a safe and effective cancer management strategy. While there is currently active research on new α-therapy regimens for prostate cancer based on the prostate-specific membrane antigen, there is emerging development of radiopharmaceutical therapy with a range of biological targets and α-emitting radioisotopes for malignancies other than the prostate cancer. This article provides a brief review of preclinical and first-in-human studies of targeted α-therapy in the cancers of brain, breast, lung, gastrointestinal, pancreas, ovary, and the urinary bladder. The data on leukemia, melanoma, myeloma, and neuroendocrine tumors will also be presented. It is anticipated that with further research the emerging role of targeted α-therapy in cancer management will be defined and validated.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine and Molecular Imaging Center, Department of Radiology, Keck School of Medicine of USC, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA, 90033, USA.
| | - Patrick M Colletti
- Division of Nuclear Medicine and Molecular Imaging Center, Department of Radiology, Keck School of Medicine of USC, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA, 90033, USA
| |
Collapse
|
18
|
A Novel Single-Step-Labeled 212Pb-CaCO 3 Microparticle for Internal Alpha Therapy: Preparation, Stability, and Preclinical Data from Mice. MATERIALS 2021; 14:ma14237130. [PMID: 34885283 PMCID: PMC8658347 DOI: 10.3390/ma14237130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
Lead-212 is recognized as a promising radionuclide for targeted alpha therapy for tumors. Many studies of 212Pb-labeling of various biomolecules through bifunctional chelators have been conducted. Another approach to exploiting the cytotoxic effect is coupling the radionuclide to a microparticle acting as a carrier vehicle, which could be used for treating disseminated cancers in body cavities. Calcium carbonate may represent a suitable material, as it is biocompatible, biodegradable, and easy to synthesize. In this work, we explored 212Pb-labeling of various CaCO3 microparticles and developed a protocol that can be straightforwardly implemented by clinicians. Vaterite microparticles stabilized by pamidronate were effective as 212Pb carriers; labeling yields of ≥98% were achieved, and 212Pb was strongly retained by the particles in an in vitro stability assessment. Moreover, the amounts of 212Pb reaching the kidneys, liver, spleen, and skeleton of mice following intraperitoneal (i.p.) administration were very low compared to i.p. injection of unbound 212Pb2+, indicating that CaCO3-bound 212Pb exhibited stability when administered intraperitoneally. Therapeutic efficacy was observed in a model of i.p. ovarian cancer for all the tested doses, ranging from 63 to 430 kBq per mouse. Lead-212-labeled CaCO3 microparticles represent a promising candidate for treating intracavitary cancers.
Collapse
|
19
|
Durand-Panteix S, Monteil J, Sage M, Garot A, Clavel M, Saidi A, Torgue J, Cogne M, Quelven I. Preclinical study of 212Pb alpha-radioimmunotherapy targeting CD20 in non-Hodgkin lymphoma. Br J Cancer 2021; 125:1657-1665. [PMID: 34671126 DOI: 10.1038/s41416-021-01585-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Despite therapeutic advances, Non-Hodgkin lymphoma (NHL) relapses can occur. The development of radioimmunotherapy (RIT) with α-emitters is an attractive alternative. In this study, we investigated the potential of α-RIT in conjunction with 212Pb-rituximab for the treatment of NHL. METHODS EL4-hCD20-Luc cells (mouse lymphoma cell line) were used for in vitro and in vivo studies. Biodistribution and efficacy studies were performed on C57BL/6 mice injected intravenously with 25 × 103 cells. RESULTS 212Pb-rituximab (0.925-7.4 kBq/mL) inhibit proliferation of EL4-hCD20-Luc cells in vitro. Biodistribution of 203/212Pb-rituximab in mice showed a significant tumour uptake and suggested that the liver, spleen, and kidneys were the organs at risk. For efficacy studies, mice were treated at either 11 days (early stage) or 20-30 days after injection of tumour cells (late stage). Treatment with 277.5 kBq 212Pb-rituximab significantly prolonged survival. Even at an advanced tumour stage, significant tumour regression occurred, with an increase in the median survival time to 28 days, compared with 9 days in the controls. CONCLUSIONS These results show the efficacy of 212Pb-rituximab in a murine syngeneic lymphoma model, in terms of significant tumour regression and increased survival, thereby highlighting the potency of α-RIT for the treatment of NHL.
Collapse
Affiliation(s)
- Stéphanie Durand-Panteix
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Jacques Monteil
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.,Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Magali Sage
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | - Armand Garot
- Nuclear Medicine Department, Limoges University Hospital, Limoges, France
| | - Marie Clavel
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France
| | | | | | - Michel Cogne
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France.
| | - Isabelle Quelven
- CNRS-UMR7276 - INSERM U1262, Contrôle de la réponse immune B et lymphoproliférations, Limoges University, Limoges, France. .,Nuclear Medicine Department, Limoges University Hospital, Limoges, France. .,ToNIC, Toulouse NeuroImaging Center - INSERM U1214, Toulouse, France.
| |
Collapse
|
20
|
Eychenne R, Chérel M, Haddad F, Guérard F, Gestin JF. Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The "Hopeful Eight". Pharmaceutics 2021; 13:pharmaceutics13060906. [PMID: 34207408 PMCID: PMC8234975 DOI: 10.3390/pharmaceutics13060906] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022] Open
Abstract
Among all existing radionuclides, only a few are of interest for therapeutic applications and more specifically for targeted alpha therapy (TAT). From this selection, actinium-225, astatine-211, bismuth-212, bismuth-213, lead-212, radium-223, terbium-149 and thorium-227 are considered as the most suitable. Despite common general features, they all have their own physical characteristics that make them singular and so promising for TAT. These radionuclides were largely studied over the last two decades, leading to a better knowledge of their production process and chemical behavior, allowing for an increasing number of biological evaluations. The aim of this review is to summarize the main properties of these eight chosen radionuclides. An overview from their availability to the resulting clinical studies, by way of chemical design and preclinical studies is discussed.
Collapse
Affiliation(s)
- Romain Eychenne
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| | - Michel Chérel
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Férid Haddad
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
- Laboratoire Subatech, UMR 6457, Université de Nantes, IMT Atlantique, CNRS, Subatech, F-44000 Nantes, France
| | - François Guérard
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
| | - Jean-François Gestin
- Université de Nantes, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Nantes—Angers (CRCINA)—UMR 1232, ERL 6001, F-44000 Nantes, France; (M.C.); (F.G.)
- Correspondence: (R.E.); (J.-F.G.)
| |
Collapse
|
21
|
Stenberg VY, Larsen RH, Ma LW, Peng Q, Juzenas P, Bruland ØS, Juzeniene A. Evaluation of the PSMA-Binding Ligand 212Pb-NG001 in Multicellular Tumour Spheroid and Mouse Models of Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094815. [PMID: 34062920 PMCID: PMC8124365 DOI: 10.3390/ijms22094815] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/12/2023] Open
Abstract
Radioligand therapy targeting the prostate-specific membrane antigen (PSMA) is rapidly evolving as a promising treatment for metastatic castration-resistant prostate cancer. The PSMA-targeting ligand p-SCN-Bn-TCMC-PSMA (NG001) labelled with 212Pb efficiently targets PSMA-positive cells in vitro and in vivo. The aim of this preclinical study was to evaluate the therapeutic potential of 212Pb-NG001 in multicellular tumour spheroid and mouse models of prostate cancer. The cytotoxic effect of 212Pb-NG001 was tested in human prostate C4-2 spheroids. Biodistribution at various time points and therapeutic effects of different activities of the radioligand were investigated in male athymic nude mice bearing C4-2 tumours, while long-term toxicity was studied in immunocompetent BALB/c mice. The radioligand induced a selective cytotoxic effect in spheroids at activity concentrations of 3–10 kBq/mL. In mice, the radioligand accumulated rapidly in tumours and was retained over 24 h, while it rapidly cleared from nontargeted tissues. Treatment with 0.25, 0.30 or 0.40 MBq of 212Pb-NG001 significantly inhibited tumour growth and improved median survival with therapeutic indexes of 1.5, 2.3 and 2.7, respectively. In BALB/c mice, no signs of long-term radiation toxicity were observed at activities of 0.05 and 0.33 MBq. The obtained results warrant clinical studies to evaluate the biodistribution, therapeutic efficacy and toxicity of 212Pb-NG001.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
- Department of Research and Development, Nucligen AS, 0379 Oslo, Norway;
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Correspondence: ; Tel.: +47-9012-8434
| | | | - Li-Wei Ma
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| | - Qian Peng
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Petras Juzenas
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| |
Collapse
|
22
|
Ahenkorah S, Cassells I, Deroose CM, Cardinaels T, Burgoyne AR, Bormans G, Ooms M, Cleeren F. Bismuth-213 for Targeted Radionuclide Therapy: From Atom to Bedside. Pharmaceutics 2021; 13:599. [PMID: 33919391 PMCID: PMC8143329 DOI: 10.3390/pharmaceutics13050599] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/17/2022] Open
Abstract
In contrast to external high energy photon or proton therapy, targeted radionuclide therapy (TRNT) is a systemic cancer treatment allowing targeted irradiation of a primary tumor and all its metastases, resulting in less collateral damage to normal tissues. The α-emitting radionuclide bismuth-213 (213Bi) has interesting properties and can be considered as a magic bullet for TRNT. The benefits and drawbacks of targeted alpha therapy with 213Bi are discussed in this review, covering the entire chain from radionuclide production to bedside. First, the radionuclide properties and production of 225Ac and its daughter 213Bi are discussed, followed by the fundamental chemical properties of bismuth. Next, an overview of available acyclic and macrocyclic bifunctional chelators for bismuth and general considerations for designing a 213Bi-radiopharmaceutical are provided. Finally, we provide an overview of preclinical and clinical studies involving 213Bi-radiopharmaceuticals, as well as the future perspectives of this promising cancer treatment option.
Collapse
Affiliation(s)
- Stephen Ahenkorah
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium; (S.A.); (I.C.); (T.C.); (A.R.B.)
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, 3000 Leuven, Belgium;
| | - Irwin Cassells
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium; (S.A.); (I.C.); (T.C.); (A.R.B.)
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, 3000 Leuven, Belgium;
| | - Christophe M. Deroose
- Nuclear Medicine Unit, University Hospitals Leuven, 3000 Leuven, Belgium;
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Thomas Cardinaels
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium; (S.A.); (I.C.); (T.C.); (A.R.B.)
- Department of Chemistry, University of Leuven, 3001 Leuven, Belgium
| | - Andrew R. Burgoyne
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium; (S.A.); (I.C.); (T.C.); (A.R.B.)
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, 3000 Leuven, Belgium;
| | - Maarten Ooms
- Institute for Nuclear Materials Science, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium; (S.A.); (I.C.); (T.C.); (A.R.B.)
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
23
|
Fiaccabrino DE, Kunz P, Radchenko V. Potential for production of medical radionuclides with on-line isotope separation at the ISAC facility at TRIUMF and particular discussion of the examples of 165Er and 155Tb. Nucl Med Biol 2021; 94-95:81-91. [PMID: 33607326 DOI: 10.1016/j.nucmedbio.2021.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/25/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
Production of medical radionuclides with ISOL facilities is a unique production method that may provide access to preclinical quantities of some rare and potent radionuclides for nuclear medicine. Particularly attention over the past years was focused on several promising candidates for Targeted Radionuclides Therapy (TRT). With this review, we provide some perspectives of using the TRIUMF ISOL facility (ISAC) to produce medical radionuclides for TRT application and highlight our current effort to collect of 165Er and 155Tb for Auger Therapy and SPECT imaging, respectively.
Collapse
Affiliation(s)
- Desiree Erika Fiaccabrino
- Life Sciences Division, TRIUMF, Vancouver, British Columbia V6T 2A3, Canada; Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Peter Kunz
- Accelerator Division, TRIUMF, Vancouver, British Columbia V6T 2A3, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, British Columbia V6T 2A3, Canada; Department of Chemistry, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada.
| |
Collapse
|
24
|
Juzeniene A, Stenberg VY, Bruland ØS, Larsen RH. Preclinical and Clinical Status of PSMA-Targeted Alpha Therapy for Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:779. [PMID: 33668474 PMCID: PMC7918517 DOI: 10.3390/cancers13040779] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Bone, lymph node, and visceral metastases are frequent in castrate-resistant prostate cancer patients. Since such patients have only a few months' survival benefit from standard therapies, there is an urgent need for new personalized therapies. The prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer and is a molecular target for imaging diagnostics and targeted radionuclide therapy (theragnostics). PSMA-targeted α therapies (PSMA-TAT) may deliver potent and local radiation more selectively to cancer cells than PSMA-targeted β- therapies. In this review, we summarize both the recent preclinical and clinical advances made in the development of PSMA-TAT, as well as the availability of therapeutic α-emitting radionuclides, the development of small molecules and antibodies targeting PSMA. Lastly, we discuss the potentials, limitations, and future perspectives of PSMA-TAT.
Collapse
Affiliation(s)
- Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway;
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379 Oslo, Norway;
- Nucligen, Ullernchausséen 64, 0379 Oslo, Norway;
- Institute for Clinical Medicine, University of Oslo, Box 1171 Blindern, 0318 Oslo, Norway;
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, Box 1171 Blindern, 0318 Oslo, Norway;
- Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | | |
Collapse
|