1
|
Ciuciulkaite I, Herrmann K, Lahner H. [Importance of peptide receptor radionuclide therapy for the management of neuroendocrine tumours]. RADIOLOGIE (HEIDELBERG, GERMANY) 2025:10.1007/s00117-025-01452-y. [PMID: 40227439 DOI: 10.1007/s00117-025-01452-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Neuroendocrine tumours (NETs) are rare, heterogeneous neoplasms that often express somatostatin receptors (SSTRs). This allows targeted peptide receptor radionuclide therapy (PRRT) for NETs. PRRT is currently indicated as second- or third-line therapy for metastatic or unresectable, progressive, SSTR-positive NETs of grade (G) 1 or 2. Adequate bone marrow reserves as well as renal and hepatic function are required for PRRT. The most commonly used radiopharmaceutical for PRRT is 177Lu-DOTA-TATE. PRRT prolongs progression-free and overall survival, reduces or stabilises tumour burden, and improves tumour symptoms and quality of life. Adverse events associated with PRRT are mostly mild and transient. Haemato- and nephrotoxicity are the most common toxicities following PRRT. The NETTER‑2 and COMPOSE trials are investigating PRRT with 177Lu-DOTA-TATE/-TOC in G2 and G3 gastroenteropancreatic NETs.
Collapse
Affiliation(s)
- I Ciuciulkaite
- Klinik für Nuklearmedizin, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland.
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - K Herrmann
- Klinik für Nuklearmedizin, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland
| | - H Lahner
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsmedizin Essen, Universität Duisburg-Essen, Essen, Deutschland
| |
Collapse
|
2
|
Belge Bilgin G, Lucien-Matteoni F, Chaudhuri AA, Orme JJ, Childs DS, Muniz M, Li GG, Chauhan PS, Lee S, Gupta S, Thorpe MP, Johnson DR, Johnson GB, Kendi AT, Sartor O. Current and future directions in theranostics for neuroendocrine prostate cancer. Cancer Treat Rev 2025; 136:102941. [PMID: 40239461 DOI: 10.1016/j.ctrv.2025.102941] [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: 02/05/2025] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) is rare at the time of initial diagnosis but much more common in patients treated with the combination of androgen deprivation therapy (ADT) and androgen receptor pathway inhibitors (ARPI) such as abiraterone and enzalutamide. NEPC is typically characterized by the loss of prostate-specific membrane antigen (PSMA) expression while exhibiting variable neuroendocrine markers. Recent advancements in nuclear medicine have provided a promising avenue for the development of molecular imaging techniques and targeted therapies tailored to NEPC. This review examines the current and future role of theranostics in the diagnosis and management of NEPC and explores potential future directions in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | - Jacob J Orme
- Department of Oncology, Mayo Clinic Rochester, MN, USA
| | | | - Miguel Muniz
- Department of Oncology, Mayo Clinic Rochester, MN, USA
| | | | | | - SeungBaek Lee
- Department of Radiology, Mayo Clinic Rochester, MN, USA
| | - Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | | | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic Rochester, MN, USA; Department of Immunology, Mayo Clinic Rochester, MN, USA
| | | | - Oliver Sartor
- Department of Radiology, Mayo Clinic Rochester, MN, USA; Department of Urology, Mayo Clinic Rochester, MN, USA; Department of Oncology, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
3
|
Ballal S, Sheokand P, Yadav MP, Roesch F, Viswanathan R, Satapathy S, Tripathi M, Moon ES, Mishra P, Rastogi S, Agarwal S, Bal C. Biodistribution and Dosimetry Evaluation of the Radiolabeled Somatostatin Receptor Antagonist 68 Ga-DATA 5m LM4 in Molecular Imaging of Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors Patients. Clin Nucl Med 2025; 50:e194-e201. [PMID: 39761436 DOI: 10.1097/rlu.0000000000005649] [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: 03/04/2025]
Abstract
PURPOSE This study aimed to assess the biodistribution and radiation dosimetry of 68 Ga-DATA 5m LM4 in patients with gastroenteropancreatic neuroendocrine tumors. PATIENTS AND METHODS Eight patients (5 females and 3 males) with various gastroenteropancreatic neuroendocrine tumors were included in the study. Each patient underwent 3 whole-body PET scans at 10, 60, and 120 minutes after receiving an IV injection of approximately 162.5 MBq of 68 Ga-DATA 5m LM4. Organs considered for dosimetric analysis included the liver, heart, spleen, kidneys, adrenal glands, and lumbar vertebrae (L2 to L4). Dosimetric calculations were performed using the OLINDA/EXM 2.2 software. RESULTS Physiological uptake of 68 Ga-DATA 5m LM4 was observed in the pituitary gland, spleen, liver, adrenal glands, and the urinary tract (kidneys and urinary bladder) for all patients. The kidneys received the highest absorbed doses at (4.77E-02 ± 1.49E-02 mSv/MBq). The mean effective dose was 2.61E-03 ± 5.99E-04 mSv/MBq. CONCLUSIONS 68 Ga-DATA 5m LM4 injection is safe and is primarily excreted through urine, delivering the highest radiation dose to the kidneys.
Collapse
Affiliation(s)
- Sanjana Ballal
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Parvind Sheokand
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Madhav P Yadav
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Frank Roesch
- Department of Chemistry-TRIGA Site, Johannes Gutenberg University, Mainz, Germany
| | - Rahul Viswanathan
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Swayamjeet Satapathy
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Madhavi Tripathi
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Euy Sung Moon
- Department of Chemistry-TRIGA Site, Johannes Gutenberg University, Mainz, Germany
| | - Prashant Mishra
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Rastogi
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Shipra Agarwal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- From the Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
4
|
Bao X, Li S, Yao S, Chen Q. Research process of PET tracers for neuroendocrine tumors diagnosis. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2025; 15:1-14. [PMID: 40124763 PMCID: PMC11929009 DOI: 10.62347/jxly1661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/14/2025] [Indexed: 03/25/2025]
Abstract
Neuroendocrine tumors (NETs) can affect several organ systems and present a variety of clinical symptoms, which are difficult to diagnose by conventional methods. Somatostatin receptor (SSTR) is a group of specific receptors expressed on the well-differentiated NET cell membrane. [68Ga]-labeled somatostatin analogues (SSAs) PET/CT, endogenous ligands targeting SSTR, is widely used in currently clinical NETs diagnosis. The dual-tracer strategy ([68Ga]Ga-SSAs + [18F]FDG) allows for a more detailed evaluation of tumor metabolism and receptor expression. The NETPET score, integrating [68Ga]Ga-SSAs PET/CT and [18F]FDG PET/CT results, enhances the accuracy of predicting treatment response and prognosis. In addition, novel isotopes ([18F]/[64Cu]) labeled SSAs and SSTR antagonists outperformed [68Ga]-SSAs in lesion detection, tumor uptake, and tumor-to-background ratio. Due to undifferentiated or dedifferentiated NETs, SSTR may not be expressed. [68Ga]Ga-Pentixafor and [18F]-FDG PET/CT are applicable for SSTR-negative NET diagnosis. [18F]-MFBG and [18F]-DOPA have a higher sensitivity for identifying non-metastatic pheochromocytoma and paraganglioma (PPGL) than other radiotracers. This review addressed NET diagnosis with conventional imaging techniques, the clinical application of novel radiotracers, and the merits and limitations of the various radiotracers.
Collapse
Affiliation(s)
- Xiangyuan Bao
- Department of PET/CT Diagnostic, Tianjin Key Lab of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General HospitalTianjin 300052, China
- The Clinical Research and Translational Center, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, Fujian, China
| | - Shuai Li
- Department of PET/CT Diagnostic, Tianjin Key Lab of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General HospitalTianjin 300052, China
| | - Shaobo Yao
- Department of PET/CT Diagnostic, Tianjin Key Lab of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General HospitalTianjin 300052, China
- The Clinical Research and Translational Center, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, Fujian, China
| | - Qiusong Chen
- Department of PET/CT Diagnostic, Tianjin Key Lab of Functional Imaging and Tianjin Institute of Radiology, Tianjin Medical University General HospitalTianjin 300052, China
| |
Collapse
|
5
|
Ebbehoj A, Iversen P, Kramer S, Stochholm K, Poulsen PL, Hjorthaug K, Søndergaard E. Positron Emission Tomography Imaging of Pheochromocytoma and Paraganglioma-18F-FDOPA vs Somatostatin Analogues. J Clin Endocrinol Metab 2025; 110:303-316. [PMID: 39468778 DOI: 10.1210/clinem/dgae764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/19/2024] [Accepted: 10/26/2024] [Indexed: 10/30/2024]
Abstract
CONTEXT Functional imaging with positron emission tomography (PET) scans is an essential part of the diagnostic workup for pheochromocytoma and paraganglioma (PPGL). The purpose of this review is to (1) provide a brief overview of functional imaging for PPGL, (2) summarize selected present and older guideline and review recommendations, and (3) conduct a literature review on the diagnostic performance of the most used PET tracers for PPGL. EVIDENCE ACQUISITION We conducted a systematic literature search in PubMed from January 2004 to August 2024 with the search string ("Pheochromocytoma" OR "Paraganglioma") AND ("Positron Emission Tomography" OR "Radionuclide Imaging" OR ("PET" AND ("FDG" OR "DOTATOC" OR "DOTANOC" OR "DOTATATE" OR "DOPA" OR "FDOPA"))). Studies involving PET scans of at least 20 individuals with PPGL or at least 5 individuals in a rare, well-defined subgroup of PPGL (eg, sympathetic or head-neck paragangliomas and specific pathogenic variants) were included. EVIDENCE SYNTHESIS Seventy studies were identified of which 21 were head-to-head comparisons of at least 2 different PET tracers [18F-fluorodihydroxyphenylalanine, fluorodihydroxyphenylalanine positron emission tomography (18F-FDOPA), 68Ga-DOTA-conjugated somatostatin analogues, 68Ga-DOTA-conjugated somatostatin analogue positron emission tomography (68Ga-SSA), and 18F-fluorodeoxyglucose]. 18F-FDOPA had higher sensitivity for pheochromocytoma compared to 68Ga-SSA and equal sensitivity for metastatic pheochromocytoma. 18F-FDOPA and 68Ga-SSA had similar sensitivity for primary non-succinate dehydrogenase subunits (SDHx) sympathetic and head-neck paraganglioma. However, 68Ga-SSA had higher sensitivity for metastatic sympathetic and head-neck paraganglioma and for SDHx-related paraganglioma. CONCLUSION 18F-FDOPA and 68Ga-SSA PET are both sensitive for localizing PPGL. However, 18F-FDOPA is the most sensitive for detecting pheochromocytoma, while 68Ga-SSA is superior to 18F-FDOPA for metastatic sympathetic and head-neck paraganglioma and SDHx-related paraganglioma.
Collapse
Affiliation(s)
- Andreas Ebbehoj
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Peter Iversen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Stine Kramer
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Kirstine Stochholm
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Per Løgstrup Poulsen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N DK-8200, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Karin Hjorthaug
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| | - Esben Søndergaard
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus N DK-8200, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N DK-8200, Denmark
| |
Collapse
|
6
|
Omweri JM, Houson HA, Lynch SE, Tekin V, Sorace AG, Lapi SE. PET imaging of 52Mn labeled DOTATATE and DOTAJR11. Sci Rep 2025; 15:2395. [PMID: 39827143 PMCID: PMC11742957 DOI: 10.1038/s41598-025-85143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025] Open
Abstract
Manganese-52 is gaining interest as an isotope for PET imaging due to its desirable decay and chemical properties for radiopharmaceutical development. Somatostatin receptor 2 (SSTR2) is significantly overexpressed by neuroendocrine tumors (NETs) and is an important target for nuclear imaging and therapy. As an agonist, [68Ga]Ga-DOTATATE has demonstrated significant internalization upon interaction with receptor ligands, whereas [68Ga]Ga-DOTA-JR11(as an antagonist) exhibits limited internalization but better pharmacokinetics and increased tumor uptake. The goal of this study was to label both DOTATATE and DOTA-JR11 peptides with 52Mn in high radiochemical yields (RCY) and sufficient specific activity. A comparison of these two compounds was performed in in vitro and in vivo studies in animals with somatostatin receptor-positive xenografts to characterize differences in cell, tumor, and tissue uptake. Radiolabeling of DOTATATE and DOTA-JR11 was carried out by combining varying concentrations of the peptides with [52Mn]MnCl2. In vitro stability of the radiotracers was determined in mouse serum. In vitro cell uptake and internalization assays were performed in SSTR2 + AR42J cells and negative controls. In vivo biodistribution and longitudinal PET imaging was evaluated in mice bearing AR42J tumors. Both [52Mn]Mn-DOTATATE and [52Mn]Mn-DOTA-JR11 showed affinity for SSTR2 in AR42J cells. However, the uptake of [52Mn]Mn-DOTATATE was higher (11.95 ± 0.71%/ mg) compared to [52Mn]Mn-DOTA-JR11 (7.31 ± 0.38%/ mg) after 2 h incubation. After 4 h incubation, 53.13 ± 1.83% of the total accumulated activity of [52Mn]Mn-DOTATATE was internalized, whereas only 20.85 ± 0.59% of the total accumulated activity of [52Mn]Mn-DOTA-JR11 was internalized. The PET images revealed similar biodistribution results, with [52Mn]Mn-DOTATATE showing a significant tumor uptake of 11.16 ± 2.97% ID/g, while [52Mn]Mn-DOTA-JR11 exhibited a lower tumor uptake of 2.11 ± 0.30% ID/g 4 h post-injection. The synthesis of both radiotracers was accomplished with high RCY and purity. The cell uptake and internalization of [52Mn]Mn-DOTATATE showed higher levels compared to [52Mn]Mn-DOTA-JR11. PET images of the radiotracers in AR42J tumor bearing mice demonstrated similar biodistribution in all organs except the tumor, with [52Mn]Mn-DOTATATE showing higher tumor uptake compared to [52Mn]Mn-DOTA-JR11. The variations in the properties of these tracers could be used to guide further imaging and treatment studies.
Collapse
Affiliation(s)
- James M Omweri
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey A Houson
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
| | - Shannon E Lynch
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Volkan Tekin
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA.
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Li Z, Ruan Q, Jiang Y, Wang Q, Yin G, Feng J, Zhang J. Current Status and Perspectives of Novel Radiopharmaceuticals with Heterologous Dual-targeted Functions: 2013-2023. J Med Chem 2024; 67:21644-21670. [PMID: 39648432 DOI: 10.1021/acs.jmedchem.4c01608] [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: 12/10/2024]
Abstract
Radiotracers provide molecular- and cellular-level information in a noninvasive manner and have become important tools for precision medicine. In particular, the successful clinical application of radioligand therapeutic (RLT) has further strengthened the role of nuclear medicine in clinical treatment. The complicated microenvironment of the lesion has rendered traditional single-targeted radiopharmaceuticals incapable of fully meeting the requirements. The design and development of dual-targeted and multitargeted radiopharmaceuticals have rapidly emerged. In recent years, significant progress has been made in the development of heterologous dual-targeted radiopharmaceuticals. This perspective aims to provide a comprehensive overview of the recent progress in these heterologous dual-targeted radiopharmaceuticals, with a special focus on the design of ligand structures, pharmacological properties, and preclinical and clinical evaluation. Furthermore, future directions are discussed from this perspective.
Collapse
Affiliation(s)
- Zuojie Li
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
8
|
Miller C, Klyuzhin I, Chaussé G, Brosch-Lenz J, Koniar H, Shi K, Rahmim A, Uribe C. Impact of cell geometry, cellular uptake region, and tumour morphology on 225Ac and 177Lu dose distributions in prostate cancer. EJNMMI Phys 2024; 11:97. [PMID: 39570450 PMCID: PMC11582247 DOI: 10.1186/s40658-024-00700-9] [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: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Radiopharmaceutical therapy with 225Ac- and 177Lu-PSMA has shown promising results for the treatment of prostate cancer. However, the distinct physical properties of alpha and beta radiation elicit varying cellular responses, which could be influenced by factors such as tumour morphology. In this study, we use simulations to examine how cell geometry, region of pharmaceutical uptake within the cell to model different internalization fractions, and the presence of tumour hypoxia and necrosis impact nucleus absorbed doses and dose heterogeneity with 225Ac and 177Lu. We also develop nucleus absorbed dose kernels for application to autoradiography images. METHODS We used the GATE Monte Carlo software to simulate three geometries of LNCaP prostate cancer cells (spherical, cubic, and ovoid) with activity of 225Ac or 177Lu internalized in the cytoplasm or bound to the extracellular membrane. Nucleus S-values were calculated for each geometry, source region, and isotope. The cell models were used to create nucleus absorbed dose kernels for each source region describing the dose to each nucleus in a cell layer, which were applied to simulated tumours composed of normoxic, hypoxic, or necrotic cancer cells to obtain dose rate maps. Absorbed doses within the tumours and dose heterogeneity were analyzed for each tumour morphology and isotope. Cell geometry made a minimal impact on S-values to the nucleus, however internalization resulted in higher nucleus doses. Applying the kernels to the simulated tumour maps showed that doses to each cell type varied between 225Ac and 177Lu depending on tumour morphology. Dose heterogeneity within tumours was slightly higher with 225Ac, however the tumour morphology made a larger impact on dose heterogeneity compared to the choice of isotope, with hypoxic and necrotic tumours having very heterogeneous dose distributions. CONCLUSIONS Cell geometry simplifications may still allow robust results in simulation studies. Furthermore, the morphology of the tumour itself may make a larger impact on treatment response compared to other variables such as ratio of internalization. Finally, nucleus absorbed dose kernels were created that could enable microdosimetric studies with autoradiography.
Collapse
Affiliation(s)
- Cassandra Miller
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Ivan Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Julia Brosch-Lenz
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Helena Koniar
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Kuangyu Shi
- Department of Nuclear Medicine, Bern University Hospital, The University of Bern, Bern, Switzerland
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
- Molecular Imaging and Therapy, BC Cancer Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Borie AM, Dromard Y, Chakraborty P, Fontanaud P, Andre EM, François A, Colson P, Muscatelli F, Guillon G, Desarménien MG, Jeanneteau F. Neuropeptide therapeutics to repress lateral septum neurons that disable sociability in an autism mouse model. Cell Rep Med 2024; 5:101781. [PMID: 39423809 PMCID: PMC11604546 DOI: 10.1016/j.xcrm.2024.101781] [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: 02/15/2024] [Revised: 06/20/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Confronting oxytocin and vasopressin deficits in autism spectrum disorders and rare syndromes brought promises and disappointments for the treatment of social disabilities. We searched downstream of oxytocin and vasopressin for targets alleviating social deficits in a mouse model of Prader-Willi syndrome and Schaaf-Yang syndrome, both associated with high prevalence of autism. We found a population of neurons in the lateral septum-activated on termination of social contacts-which oxytocin and vasopressin inhibit as per degree of peer affiliation. These are somatostatin neurons expressing oxytocin receptors coupled to GABA-B signaling, which are inhibited via GABA-A channels by vasopressin-excited GABA neurons. Loss of oxytocin or vasopressin signaling recapitulated the disease phenotype. By contrast, deactivation of somatostatin neurons or receptor signaling alleviated social deficits of disease models by increasing the duration of contacts with mates and strangers. These findings provide new insights into the treatment framework of social disabilities in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Amélie M Borie
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Yann Dromard
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Prabahan Chakraborty
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Pierre Fontanaud
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Emilie M Andre
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France; Département de Maieutique, University of Montpellier, 34090 Montpellier, France
| | - Amaury François
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Pascal Colson
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France; Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital, Montpellier 34090 Montpellier, France
| | - Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée, INSERM, University of Aix-Marseille, 13273 Marseille, France
| | - Gilles Guillon
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Michel G Desarménien
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France.
| |
Collapse
|
10
|
Busslinger SD, Mapanao AK, Kegler K, Bernhardt P, Flühmann F, Fricke J, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Comparison of the tolerability of 161Tb- and 177Lu-labeled somatostatin analogues in the preclinical setting. Eur J Nucl Med Mol Imaging 2024; 51:4049-4061. [PMID: 39046521 PMCID: PMC11527941 DOI: 10.1007/s00259-024-06827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/30/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE [177Lu]Lu-DOTATATE is an established somatostatin receptor (SSTR) agonist for the treatment of metastasized neuroendocrine neoplasms, while the SSTR antagonist [177Lu]Lu-DOTA-LM3 has only scarcely been employed in clinics. Impressive preclinical data obtained with [161Tb]Tb-DOTA-LM3 in tumor-bearing mice indicated the potential of terbium-161 as an alternative to lutetium-177. The aim of the present study was to compare the tolerability of 161Tb- and 177Lu-based DOTA-LM3 and DOTATATE in immunocompetent mice. METHODS Dosimetry calculations were performed based on biodistribution data of the radiopeptides in immunocompetent mice. Treatment-related effects on blood cell counts were assessed on Days 10, 28 and 56 after application of [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE at 20 MBq per mouse. These radiopeptides were also applied at 100 MBq per mouse and the effects compared to those observed after application of the 177Lu-labeled counterparts. Bone marrow smears, blood plasma parameters and organ histology were assessed at the end of the study. RESULTS The absorbed organ dose was commonly higher for the SSTR antagonist than for the SSTR agonist and for terbium-161 over lutetium-177. Application of a therapeutic activity level of 20 MBq [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE was well tolerated without major hematological changes. The injection of 100 MBq of the 161Tb- and 177Lu-based somatostatin analogues affected the blood cell counts, however. The lymphocytes were 40-50% lower in treated mice compared to the untreated controls on Day 10 irrespective of the radionuclide employed. At the same timepoint, thrombocyte and erythrocyte counts were 30-50% and 6-12% lower, respectively, after administration of the SSTR antagonist (p < 0.05) while changes were less pronounced in mice injected with the SSTR agonist. All blood cell counts were in the normal range on Day 56. Histological analyses revealed minimal abnormalities in the kidneys, liver and spleen of treated mice. No correlation was observed between the organ dose and frequency of the occurrence of abnormalities. CONCLUSION Hematologic changes were more pronounced in mice treated with the SSTR antagonist than in those treated with the SSTR agonist. Despite the increased absorbed dose delivered by terbium-161 over lutetium-177, [161Tb]Tb-DOTA-LM3 and [161Tb]Tb-DOTATATE should be safe at activity levels that are recommended for their respective 177Lu-based analogues.
Collapse
Affiliation(s)
- Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | | | - Peter Bernhardt
- Department of Medical Radiation Sciences, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Department of Medical Physics and Biomedical Engineering (MFT), Sahlgrenska University Hospital, Gothenburg, 41345, Sweden
| | - Fabienne Flühmann
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Julia Fricke
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, 0240, South Africa
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
11
|
Zhou X, Shi B, Huang G, Liu J, Wei W. Trends in cancer imaging. Trends Cancer 2024; 10:1023-1037. [PMID: 39232974 DOI: 10.1016/j.trecan.2024.08.006] [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: 05/25/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
Molecular imaging of cancer is a collaborative endeavor, uniting scientists and physicians from diverse fields. Such collaboration is actively developing and translating cutting-edge molecular imaging approaches to enhance the diagnostic landscape of human malignancies. The advent of positron emission tomography (PET) and PET imaging tracers has realized non-invasive target annotation and tumor characterization at the molecular level. In surgical procedures, novel imaging techniques, such as fluorescence or Cherenkov luminescence, help identify tumors and enhance surgical precision. Simultaneously, progress in imaging equipment, innovative algorithms, and artificial intelligence has opened avenues for next-generation cancer screening and imaging, augmenting the efficiency and accuracy of cancer diagnosis. In this review, we provide a panorama of molecular cancer imaging and ongoing developments in the field.
Collapse
Affiliation(s)
- Xinyuan Zhou
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Binyu Shi
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
12
|
Huang J, Zhang X, Liu Q, Gong F, Huang Y, Huang S, Fu L, Tang G. 68Ga/ 177Lu-Labeled Theranostic Pair for Targeting Fibroblast Activation Protein with Improved Tumor Uptake and Retention. J Med Chem 2024; 67:17785-17795. [PMID: 39321030 DOI: 10.1021/acs.jmedchem.4c01812] [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: 09/27/2024]
Abstract
Fibroblast activation protein (FAP) is specifically expressed on cancer-associated fibroblasts in over 90% of tumors and is considered a promising target for cancer theranostics. Here, we developed a novel tracer, DOTA-FAPT, and labeled it with gallium-68 and lutetium-177 as a theranostic pair. [68Ga]Ga/[177Lu]Lu-FAPT exhibited high stability and hydrophilicity, as well as strong affinity to the FAP target. Micro-PET/CT imaging revealed that [68Ga]Ga-FAPT exhibited significantly increased uptake in tumors and extended retention in A549-FAP and U87MG tumor xenografts as compared to [68Ga]Ga-FAPI-04, demonstrating favorable pharmacokinetic characteristics in vivo. Therapeutic studies showed that [177Lu]Lu-FAPT had higher tumor accumulation compared to [177Lu]Lu-FAPI-04, leading to stronger tumor growth inhibition. The first-in-human evaluation also revealed that [68Ga]Ga-FAPT has good in vivo distribution and superior diagnostic efficacy on primary and lymph node metastases in a patient with lung cancer. Our encouraging results suggest that 68Ga/177Lu-labeled DOTA-FAPT is a theranostic pair with broad application prospect.
Collapse
Affiliation(s)
- Jiawen Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xiaojun Zhang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Qingxing Liu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Fengping Gong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yanchao Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Shun Huang
- Department of Nuclear Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523059, P. R. China
| | - Lilan Fu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Ganghua Tang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| |
Collapse
|
13
|
Aloj L, Mansi R, De Luca S, Accardo A, Tesauro D, Morelli G. Radiolabeled peptides and their expanding role in clinical imaging and targeted cancer therapy. J Pept Sci 2024; 30:e3607. [PMID: 38710638 DOI: 10.1002/psc.3607] [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: 02/19/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024]
Abstract
There is an expanding body of evidence showing that synthetic peptides in combination with radioactive isotopes can be utilized for medical purposes. This area is of particular interest in oncology where applications in diagnosis and therapy are at different stages of development. We review the contributions in this area by the group originally founded by Carlo Pedone in Naples many years ago. We highlight the work of this group in the context of other developments in this area, focusing on three biologically relevant receptor systems: somatostatin, gastrin-releasing peptide, and cholecystokinin-2/gastrin receptors. We focus on key milestones, state of the art, and challenges in this area of research as well as the current and future outlook for expanding clinical applications.
Collapse
Affiliation(s)
- Luigi Aloj
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Stefania De Luca
- Institute of Biostructures and Bioimaging (IBB), CNR, Naples, Italy
| | - Antonella Accardo
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Diego Tesauro
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| | - Giancarlo Morelli
- Department of Pharmacy and CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", Naples, Italy
| |
Collapse
|
14
|
Liu M, Ren C, Zhang H, Zhang Y, Huang Z, Jia R, Cheng Y, Bai C, Xu Q, Zhu W, Huo L. Evaluation of the safety, biodistribution, dosimetry of [ 18F]AlF-NOTA-LM3 and head-to-head comparison with [ 68Ga]Ga-DOTATATE in patients with well-differentiated neuroendocrine tumors: an interim analysis of a prospective trial. Eur J Nucl Med Mol Imaging 2024; 51:3719-3730. [PMID: 38878175 DOI: 10.1007/s00259-024-06790-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/04/2024] [Indexed: 10/02/2024]
Abstract
PURPOSE 18F-labelled somatostatin receptor (SSTR) analogs offer several advantages over 68Ga in terms of yield, cost, spatial resolution and detection rate. This study presents an interim analysis of a prospective trial designed to assess the safety, biodistribution and dosimetry of [18F]AlF-NOTA-LM3, and compare its diagnostic efficacy and clinical management outcomes with [68Ga]Ga-DOTATATE or [68Ga]Ga-NODAGA-LM3 in patients with well-differentiated NETs. METHODS Twenty-one patients with histologically confirmed well-differentiated neuroendocrine tumors (G1 and G2) were prospectively recruited. The first eight patients underwent serial PET scans at 5, 15, 30, 45, 60, and 120 min after [18F]AlF-NOTA-LM3 injection to assess biodistribution and dosimetry. The remaining patients underwent whole-body PET/CT scans. [18F]AlF-NOTA-LM3 and [68Ga]Ga-DOTATATE PET/CT were done within a week, with a minimum 24-hour interval between the two scans. Focal uptake above the surrounding background activity and could not be explained by physiologic uptake was considered lesions of NETs. Lesion number, tumor uptake, and tumor-to-background ratio (TBR) were compared. In patients with discrepant findings, the size of the smallest lesions (measured on coregistered CT) detected on [68Ga]Ga-DOTATATE and [18F]AlF-NOTA-LM3 was compared. RESULTS [18F]AlF-NOTA-LM3 was safe and well-tolerated. Physiological uptake of [18F]AlF-NOTA-LM3 was significantly lower than that of [68Ga]Ga-DOTATATE in abdominal organs and bone marrow, but higher in blood pool and lung. The mean effective dose was 0.024 ± 0.014 mSv/MBq. [18F]AlF-NOTA-LM3 detected significantly more liver lesions (457 vs. 291, P = 0.006) and lymph node lesions (30 vs. 22, P = 0.011) compared to [68Ga]Ga-DOTATATE. The tumor uptake was comparable, but TBR was significantly higher with [18F]AlF-NOTA-LM3 for lesions from all sites except for the duodenum. The size of the minimum liver lesions (0.54 ± 0.15 vs. 1.01 ± 0.49, P<0.001) and lymph node lesions (0.50 ± 0.19 vs. 1.26 ± 0.86, P = 0.024) detected on [18F]ALF-NOTA-LM3 were significantly smaller than those detected on [68Ga]Ga-DOTATATE. CONCLUSION [18F]AlF-NOTA-LM3 shows favorable biodistribution, higher spatial resolution and superior performance than [68Ga]Ga-DOTATATE in detecting liver and lymph node metastases, with higher TBR. Notably, it is the first SSTR analog to show superiority in detecting lymph node lesions when compared to [68Ga]Ga-DOTATATE. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT06056362.
Collapse
Affiliation(s)
- Meixi Liu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chao Ren
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Haiqiong Zhang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuwei Zhang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhenghai Huang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ru Jia
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, China
| | - Yuejuan Cheng
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, Beijing, 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, Beijing, 100730, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, Beijing, 100730, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
15
|
Periferakis A, Tsigas G, Periferakis AT, Tone CM, Hemes DA, Periferakis K, Troumpata L, Badarau IA, Scheau C, Caruntu A, Savulescu-Fiedler I, Caruntu C, Scheau AE. Agonists, Antagonists and Receptors of Somatostatin: Pathophysiological and Therapeutical Implications in Neoplasias. Curr Issues Mol Biol 2024; 46:9721-9759. [PMID: 39329930 PMCID: PMC11430067 DOI: 10.3390/cimb46090578] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Somatostatin is a peptide that plays a variety of roles such as neurotransmitter and endocrine regulator; its actions as a cell regulator in various tissues of the human body are represented mainly by inhibitory effects, and it shows potent activity despite its physiological low concentrations. Somatostatin binds to specific receptors, called somatostatin receptors (SSTRs), which have different tissue distributions and associated signaling pathways. The expression of SSTRs can be altered in various conditions, including tumors; therefore, they can be used as biomarkers for cancer cell susceptibility to certain pharmacological agents and can provide prognostic information regarding disease evolution. Moreover, based on the affinity of somatostatin analogs for the different types of SSTRs, the therapeutic range includes conditions such as tumors, acromegaly, post-prandial hypotension, hyperinsulinism, and many more. On the other hand, a number of somatostatin antagonists may prove useful in certain medical settings, based on their differential affinity for SSTRs. The aim of this review is to present in detail the principal characteristics of all five SSTRs and to provide an overview of the associated therapeutic potential in neoplasias.
Collapse
Affiliation(s)
- Argyrios Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
| | - Georgios Tsigas
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Carla Mihaela Tone
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Daria Alexandra Hemes
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs, 17236 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, "Foisor" Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, The "Carol Davila" Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, "Titu Maiorescu" University, 031593 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
16
|
Zhao X, Jakobsson V, Tao Y, Zhao T, Wang J, Khong PL, Chen X, Zhang J. Targeted Radionuclide Therapy in Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39042829 DOI: 10.1021/acsami.4c07850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yucen Tao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jingyan Wang
- Xiamen University, School of Public Health, Xiang'an South Road, Xiamen 361102, China
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
17
|
Cai K, Zhu Y, Zheng Y, Wang H, Qian Y. B2R-Targeting Radiotracer for PET/MR Imaging of Hepatocellular Carcinoma and Guiding Anti-B2R Therapy. ACS Med Chem Lett 2024; 15:1080-1087. [PMID: 39015273 PMCID: PMC11247633 DOI: 10.1021/acsmedchemlett.4c00155] [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: 04/08/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
The bradykinin B2 receptor (B2R) is overexpressed in a wide variety of tumors and is a well-defined target for tumor imaging and therapy. The hybrid positron emission tomography/magnetic resonance imaging (PET/MRI) scanner is considered a noninvasive and advanced instrument for precise tumor imaging. In this work, we developed a novel B2R-targeting radiotracer, 68Ga-DOTA-icatibant, for quantifying B2R expression. 68Ga-DOTA-icatibant showed high stability, fast clearance and specific binding to B2R. PET/MR imaging revealed excellent tumor accumulation, and the uptake in tumors could be blocked by DOTA-icatibant. Icatibant-mediated anti-B2R therapy downregulated B2R expression in tumor cells and inhibited the growth of HepG2 tumors, and the decrease in tumor uptake was monitored by timely PET/MR imaging. Hematoxylin and eosin (H&E) and immunohistochemical staining results further demonstrated that the efficacy of anti-B2R could be accurately monitored with the developed PET/MR imaging radiotracer. 68Ga-DOTA-icatibant can be utilized to noninvasively determine B2R expression and dynamically and sensitively monitor the efficacy of anti-B2R therapy.
Collapse
Affiliation(s)
- Ke Cai
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yunzhu Zhu
- Department
of Infectious Diseases, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yifan Zheng
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Hui Wang
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| | - Yinfeng Qian
- Department
of Nuclear Medicine, The First Affiliated
Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
- Department
of Radiology, The First Affiliated Hospital
of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui Province, China
| |
Collapse
|
18
|
Massironi S, Franchina M, Ippolito D, Elisei F, Falco O, Maino C, Pagni F, Elvevi A, Guerra L, Invernizzi P. Improvements and future perspective in diagnostic tools for neuroendocrine neoplasms. Expert Rev Endocrinol Metab 2024; 19:349-366. [PMID: 38836602 DOI: 10.1080/17446651.2024.2363537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Neuroendocrine neoplasms (NENs) represent a complex group of tumors arising from neuroendocrine cells, characterized by heterogeneous behavior and challenging diagnostics. Despite advancements in medical technology, NENs present a major challenge in early detection, often leading to delayed diagnosis and variable outcomes. This review aims to provide an in-depth analysis of current diagnostic methods as well as the evolving and future directions of diagnostic strategies for NENs. AREA COVERED The review extensively covers the evolution of diagnostic tools for NENs, from traditional imaging and biochemical tests to advanced genomic profiling and next-generation sequencing. The emerging role of technologies such as artificial intelligence, machine learning, and liquid biopsies could improve diagnostic precision, as could the integration of imaging modalities such as positron emission tomography (PET)/magnetic resonance imaging (MRI) hybrids and innovative radiotracers. EXPERT OPINION Despite progress, there is still a significant gap in the early diagnosis of NENs. Bridging this diagnostic gap and integrating advanced technologies and precision medicine are crucial to improving patient outcomes. However, challenges such as low clinical awareness, limited possibility of noninvasive diagnostic tools and funding limitations for rare diseases like NENs are acknowledged.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Marianna Franchina
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Davide Ippolito
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Federica Elisei
- Division of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Olga Falco
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Cesare Maino
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Division of Pathology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandra Elvevi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Luca Guerra
- Division of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
19
|
Sakellis C, Jacene HA. Neuroendocrine Tumors: Diagnostics. PET Clin 2024; 19:325-339. [PMID: 38714399 DOI: 10.1016/j.cpet.2024.03.008] [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: 05/09/2024]
Abstract
Neuroendocrine neoplasms (NEN) are rare tumors arising from neuroendocrine cells. NEN are ideally suited for a theragnostic approach due to their specific expression of somatostatin receptors (SSTR). SSTR imaging of NEN dates back to the 1980s, but has evolved recently due to the introduction of more sensitive SSTR PET radiotracers. SSTR PET is a primary imaging modality for identifying NEN and characterizing SSTR expression. SSTR PET is complementary to anatomic imaging for assessing tumor response to treatment. SSTR PET is mandated to determine eligibility for peptide receptor radionuclide therapy. Here, the role of imaging to aid management of NEN is reviewed.
Collapse
Affiliation(s)
- Christopher Sakellis
- Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Avenue, DL198, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02215, USA
| | - Heather A Jacene
- Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Avenue, DL198, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02215, USA.
| |
Collapse
|
20
|
Fang J, Wang X, Meng L, Zhang J, Zhuang R, Li Y, Zhang X, Guo Z. Preclinical Evaluation of 131I/ 18F-Labeled Covalent Small-Molecule Inhibitors for STING Status Imaging. ACS Pharmacol Transl Sci 2024; 7:1783-1794. [PMID: 38898942 PMCID: PMC11184601 DOI: 10.1021/acsptsci.3c00398] [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: 12/31/2023] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 06/21/2024]
Abstract
The stimulator of interferon genes (STING) is a vital protein to the immune surveillance of the tumor microenvironment. In this study, we develop novel inhibitor-based radioligands and evaluate their feasibility for noninvasive visualization of STING expression in tumor-bearing mice. Analogous compounds to STING inhibitors C170 and C176 were synthesized and labeled with 131I and 18F to attain [131I]I-NFIP and [18F]F-NFEP, respectively. The radiosynthesis was achieved with high radiochemical purity (>95%) and molar activity (28.56-48.89 GBq/μmol). The affinity and specificity of tracers were assessed through cell uptake and docking experiments, demonstrating that [131I]I-NFIP exhibited high specificity for STING, with a cell-based IC50 value of 7.56 nM. Small-animal PET/SPECT imaging and biodistribution studies in tumor-bearing mice models were performed to verify the tracers' pharmacokinetics and tumor-targeting capabilities (n = 3/group). SPECT imaging demonstrated that [131I]I-NFIP rapidly accumulated in the Panc02 tumor quickly at 30 min post-injection, with a tumor-to-muscle (T/M) ratio of 2.03 ± 0.30. This ratio significantly decreased in the blocking group (1.10 ± 0.14, **P < 0.01, n = 3). Furthermore, tumor uptake and the T/M ratio of [131I]I-NFIP were positively associated with STING expression. In summary, [131I]I-NFIP is the first STING-specific inhibitor-based radioligand offering the potential for visualizing STING status in tumors.
Collapse
Affiliation(s)
- Jianyang Fang
- State
Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular
Imaging and Translational Medicine, Xiang An Biomedicine Laboratory,
School of Public Health, Xiamen University, 4221-116 Xiang’An South Rd, Xiamen 361102, China
| | - Xiaobo Wang
- Department
of Nuclear Medicine, Xijing Hospital, Fourth
Military Medical University, Xi’an 71003, China
| | - Lingxin Meng
- State
Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular
Imaging and Translational Medicine, Xiang An Biomedicine Laboratory,
School of Public Health, Xiamen University, 4221-116 Xiang’An South Rd, Xiamen 361102, China
| | - Jingru Zhang
- State
Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular
Imaging and Translational Medicine, Xiang An Biomedicine Laboratory,
School of Public Health, Xiamen University, 4221-116 Xiang’An South Rd, Xiamen 361102, China
| | - Rongqiang Zhuang
- State
Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular
Imaging and Translational Medicine, Xiang An Biomedicine Laboratory,
School of Public Health, Xiamen University, 4221-116 Xiang’An South Rd, Xiamen 361102, China
| | - Yesen Li
- Department
of Nuclear Medicine and Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Xianzhong Zhang
- Theranostics
and Translational Research Center, Institute of Clinical Medicine,
Department of Nuclear Medicine, Peking Union
Medical College Hospital, Chinese Academy of Medical Sciences and
Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhide Guo
- State
Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular
Imaging and Translational Medicine, Xiang An Biomedicine Laboratory,
School of Public Health, Xiamen University, 4221-116 Xiang’An South Rd, Xiamen 361102, China
| |
Collapse
|
21
|
Liu M, Cheng Y, Bai C, Zhao H, Jia R, Chen J, Zhu W, Huo L. Gallium-68 labeled somatostatin receptor antagonist PET/CT in over 500 patients with neuroendocrine neoplasms: experience from a single center in China. Eur J Nucl Med Mol Imaging 2024; 51:2002-2011. [PMID: 38337073 DOI: 10.1007/s00259-024-06639-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE Somatostatin receptor antagonists have shown promising performance for imaging neuroendocrine neoplasms. However, there is a lack of studies exploring the diagnostic performance of SSTR antagonists or comparing them with agonists in a large cohort of patients with NENs. This study aimed to retrospectively review all SSTR antagonist PET/CT scans conducted at Peking Union Medical College Hospital since November 2018 in patients with confirmed or suspected NENs. METHODS Four types of SSTR antagonists were utilized, including [68Ga]Ga-NODAGA-LM3, [68Ga]Ga-DOTA-LM3, [68Ga]Ga-NODAGA-JR11, and [68Ga]Ga-DOTA-JR11. The reference standard was based on a combination of histopathology, clinical evaluation, imaging results, and follow-up. Patient-based sensitivity, specificity, and accuracy were evaluated. The SUVmax and tumor-to-liver ratio (TLR) of the hottest lesions was recorded and compared between antagonists and [68Ga]Ga-DOTATATE. RESULTS A total of 622 antagonist scans from 549 patients were included in the analysis. The patient-level sensitivity, specificity, and accuracy of antagonist imaging (all tracers combined) were 91.0% (443/487), 91.9% (57/62), and 91.1% (500/549), respectively. In 181 patients with a comparative [68Ga]Ga-DOTATATE PET/CT scan, the patient-level sensitivity, specificity, and accuracy were 87.5% (147/168), 76.9% (10/13), and 86.7% (157/181), respectively. For the hottest lesions, SSTR antagonists all tracers combined demonstrated an overall comparable SUVmax to [68Ga]Ga-DOTATATE (40.1 ± 32.5 vs. 39.4 ± 23.8, p = 0.772). While [68Ga]Ga-NODAGA-LM3 showed significantly higher uptake than [68Ga]Ga-DOTATATE (57.4 ± 38.5 vs. 40.0 ± 22.8, p<0.001), [68Ga]Ga-NODAGA-JR11 (39.7 ± 26.5 vs. 34.3 ± 23.9, p = 0.108) and [68Ga]Ga-DOTA-LM3 (38.9 ± 32.1 vs. 37.2 ± 22.1, p = 0.858) showed comparable uptake to [68Ga]Ga-DOTATATE, and [68Ga]Ga-DOTA-JR11 showed lower uptake (28.9 ± 26.1 vs. 44.0 ± 25.7, p = 0.001). All antagonists exhibited significantly higher TLR than [68Ga]Ga-DOTATATE (12.1 ± 10.8 vs. 5.2 ± 4.5, p<0.001). CONCLUSION Gallium-68 labeled SSTR antagonists could serve as alternatives to SSTR agonists for imaging of NENs. Among various antagonists, [68Ga]Ga-NODAGA-LM3 seems to have the best imaging profile.
Collapse
Affiliation(s)
- Meixi Liu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuejuan Cheng
- Department of Oncology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ru Jia
- Department of Gastrointestinal Oncology, the fifth Medical Center, General Hospital of PLA, Beijing, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
22
|
Pougoue Ketchemen J, Njotu FN, Babeker H, Ahenkorah S, Tikum AF, Nwangele E, Henning N, Cleeren F, Fonge H. Effectiveness of [ 67Cu]Cu-trastuzumab as a theranostic against HER2-positive breast cancer. Eur J Nucl Med Mol Imaging 2024; 51:2070-2084. [PMID: 38376808 DOI: 10.1007/s00259-024-06648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE To evaluate the imaging and therapeutic properties (theranostic) of 67Cu-labeled anti-human epidermal growth factor receptor II (HER2) monoclonal antibody trastuzumab against HER2-positive breast cancer (BC). METHODS We conjugated trastuzumab with p-SCN-Bn-NOTA, 3p-C-NETA-NCS, or p-SCN-Bn-DOTA, and radiolabeled with [67Cu]CuCl2. Immunoconjugate internalization was evaluated in BT-474, JIMT-1 and MCF-7 BC cells. In vitro stability was studied in human serum (HS) and Phosphate Buffered Saline (PBS). Flow cytometry, radioligand binding and immunoreactive fraction assays were carried out. ImmunoSPECT imaging of [67Cu]Cu-NOTA-trastuzumab was done in mice bearing BT-474, JIMT-1 and MCF-7 xenografts. Pharmacokinetic was studied in healthy Balb/c mice while dosimetry was done in both healthy Balb/c and in athymic nude mice bearing JIMT-1 xenograft. The therapeutic effectiveness of [67Cu]Cu-NOTA-trastuzumab was evaluated in mice bearing BT-474 and JIMT-1 xenografts after a single intravenous (i.v.) injection of ~ 16.8 MBq. RESULTS Pure immunoconjugates and radioimmunoconjugates (> 95%) were obtained. Internalization was HER2 density-dependent with highest internalization observed with NOTA-trastuzumab. After 5 days, in vitro stabilities were 97 ± 1.7%, 31 ± 6.2%, and 28 ± 4% in HS, and 79 ± 3.5%, 94 ± 1.2%, and 86 ± 2.3% in PBS for [67Cu]Cu-NOTA-trastuzumab, [67Cu]Cu-3p-C-NETA-trastuzumab and [67Cu]Cu-DOTA-trastuzumab, respectively. [67Cu]Cu-NOTA-trastuzumab was chosen for further evaluation. BT-474 flow cytometry showed low KD, 8.2 ± 0.2 nM for trastuzumab vs 26.5 ± 1.6 nM for NOTA-trastuzumab. There were 2.9 NOTA molecules per trastuzumab molecule. Radioligand binding assay showed a low KD of 2.1 ± 0.4 nM and immunoreactive fraction of 69.3 ± 0.9. Highest uptake of [67Cu]Cu-NOTA-trastuzumab was observed in JIMT-1 (33.9 ± 5.5% IA/g) and BT-474 (33.1 ± 10.6% IA/g) xenograft at 120 h post injection (p.i.). Effectiveness of the radioimmunoconjugate was also expressed as percent tumor growth inhibition (%TGI). [67Cu]Cu-NOTA-trastuzumab was more effective than trastuzumab against BT-474 xenografts (78% vs 54% TGI after 28 days), and JIMT-1 xenografts (90% vs 23% TGI after 19 days). Mean survival of [67Cu]Cu-NOTA-trastuzumab, trastuzumab and saline treated groups were > 90, 77 and 72 days for BT-474 xenografts, while that of JIMT-1 were 78, 24, and 20 days, respectively. CONCLUSION [67Cu]Cu-NOTA-trastuzumab is a promising theranostic agent against HER2-positive BC.
Collapse
Affiliation(s)
- Jessica Pougoue Ketchemen
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
| | - Fabrice Ngoh Njotu
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
- Department of Pathology and Lab. Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK, S7N 5A2, Canada
| | - Hanan Babeker
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
- Department of Pathology and Lab. Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK, S7N 5A2, Canada
| | - Stephen Ahenkorah
- NURA Research Group, Belgian Nuclear Research Center (SCK CEN), Mol, Belgium
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, Leuven, Belgium
| | - Anjong Florence Tikum
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
| | - Emmanuel Nwangele
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
- Department of Pathology and Lab. Medicine, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK, S7N 5A2, Canada
| | - Nikita Henning
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, Leuven, Belgium
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK, S7N 0W8, Canada.
- Department of Medical Imaging, Royal University Hospital Saskatoon, Saskatoon, SK, S7N 0W8, Canada.
| |
Collapse
|
23
|
Kleynhans J, Ebenhan T, Cleeren F, Sathekge MM. Can current preclinical strategies for radiopharmaceutical development meet the needs of targeted alpha therapy? Eur J Nucl Med Mol Imaging 2024; 51:1965-1980. [PMID: 38676735 PMCID: PMC11139742 DOI: 10.1007/s00259-024-06719-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Preclinical studies are essential for effectively evaluating TAT radiopharmaceuticals. Given the current suboptimal supply chain of these radionuclides, animal studies must be refined to produce the most translatable TAT agents with the greatest clinical potential. Vector design is pivotal, emphasizing harmonious physical and biological characteristics among the vector, target, and radionuclide. The scarcity of alpha-emitting radionuclides remains a significant consideration. Actinium-225 and lead-212 appear as the most readily available radionuclides at this stage. Available animal models for researchers encompass xenografts, allografts, and PDX (patient-derived xenograft) models. Emerging strategies for imaging alpha-emitters are also briefly explored. Ultimately, preclinical research must address two critical aspects: (1) offering valuable insights into balancing safety and efficacy, and (2) providing guidance on the optimal dosing of the TAT agent.
Collapse
Affiliation(s)
- Janke Kleynhans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Thomas Ebenhan
- Department of Nuclear Medicine, University of Pretoria, and Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Mike Machaba Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa.
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, 0001, South Africa.
| |
Collapse
|
24
|
AghaAmiri S, Ghosh SC, Hernandez Vargas S, Halperin DM, Azhdarinia A. Somatostatin Receptor Subtype-2 Targeting System for Specific Delivery of Temozolomide. J Med Chem 2024; 67:2425-2437. [PMID: 38346097 PMCID: PMC10896214 DOI: 10.1021/acs.jmedchem.3c00223] [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] [Received: 02/08/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 02/23/2024]
Abstract
Temozolomide (TMZ) is a DNA alkylating agent that produces objective responses in patients with neuroendocrine tumors (NETs) when the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) is inactivated. At high doses, TMZ therapy exhausts MGMT activity but also produces dose-limiting toxicities. To reduce off-target effects, we converted the clinically approved radiotracer 68Ga-DOTA-TOC into a peptide-drug conjugate (PDC) for targeted delivery of TMZ to somatostatin receptor subtype-2 (SSTR2)-positive tumor cells. We used an integrated radiolabeling strategy for direct quantitative assessment of receptor binding, pharmacokinetics, and tissue biodistribution. In vitro studies revealed selective binding to SSTR2-positive cells with high affinity (5.98 ± 0.96 nmol/L), internalization, receptor-dependent DNA damage, cytotoxicity, and MGMT depletion. Imaging and biodistribution analysis showed preferential accumulation of the PDC in receptor-positive tumors and high renal clearance. This study identified a trackable SSTR2-targeting system for TMZ delivery and utilizes a modular design that could be broadly applied in PDC development.
Collapse
Affiliation(s)
- Solmaz AghaAmiri
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, 1881 East Road, 3SCR6.4680, Houston, Texas 77054, United States
| | - Sukhen C. Ghosh
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, 1881 East Road, 3SCR6.4680, Houston, Texas 77054, United States
| | - Servando Hernandez Vargas
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, 1881 East Road, 3SCR6.4680, Houston, Texas 77054, United States
| | - Daniel M. Halperin
- Department
of Gastrointestinal Medical Oncology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Ali Azhdarinia
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, 1881 East Road, 3SCR6.4680, Houston, Texas 77054, United States
| |
Collapse
|
25
|
Al-Toubah T, Strosberg J, Hallanger-Johnson J, El-Haddad G. Targeted radionuclide therapy in endocrine-related cancers: advances in the last decade. Front Endocrinol (Lausanne) 2023; 14:1187870. [PMID: 38053729 PMCID: PMC10694449 DOI: 10.3389/fendo.2023.1187870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Targeted radionuclide therapy plays an increasingly important role in managing endocrine-related tumors and significantly advances the therapeutic landscape for patients with these diseases. With increasing FDA-approved therapies and advances in the field, come an increased knowledge of the potential for long-term toxicities associated with these therapies and the field must develop new strategies to increase potency and efficacy while individualizing the selection of patients to those most likely to respond to treatment. Novel agents and modalities of therapy are also being explored. This review will discuss the current landscape and describe the avenues for growth in the field currently being explored.
Collapse
Affiliation(s)
- Taymeyah Al-Toubah
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Julie Hallanger-Johnson
- Department of Head and Neck - Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Ghassan El-Haddad
- Department of Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Nuclear Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
26
|
Opalinska M, Lezaic L, Decristoforo C, Kolenc P, Mikolajczak R, Studen A, Simoncic U, Virgolini I, Trofimiuk-Muldner M, Garnuszek P, Rangger C, Fani M, Glowa B, Skorkiewicz K, Hubalewska-Dydejczyk A. Comparison of 99mTc radiolabeled somatostatin antagonist with [ 68 Ga]Ga-DOTA-TATE in a patient with advanced neuroendocrine tumor. Eur J Nucl Med Mol Imaging 2023; 50:4110-4111. [PMID: 37452871 PMCID: PMC10611881 DOI: 10.1007/s00259-023-06335-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Affiliation(s)
- Marta Opalinska
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Krakow, Poland
| | - Luka Lezaic
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Petra Kolenc
- Department of Nuclear Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Renata Mikolajczak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Otwock, Poland
| | - Andrej Studen
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
- Jožef Stefan Institute, Ljubljana, Slovenia
| | - Urban Simoncic
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
- Jožef Stefan Institute, Ljubljana, Slovenia
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | - Piotr Garnuszek
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Otwock, Poland
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
27
|
DaSilva J, Decristoforo C, Mach RH, Bormans G, Carlucci G, Al-Qahtani M, Duatti A, Gee AD, Szymanski W, Rubow S, Hendrikx J, Yang X, Jia H, Zhang J, Caravan P, Yang H, Zeevaart JR, Rodriquez MA, Oliveira RS, Zubillaga M, Sakr T, Spreckelmeyer S. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2023; 8:35. [PMID: 37889361 PMCID: PMC10611660 DOI: 10.1186/s41181-023-00218-y] [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: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] 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 21 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)
| | | | | | | | | | | | | | | | | | | | | | - Xing Yang
- Peking University First Hospital, Beijing, China
| | | | | | - Peter Caravan
- Massuchusetts General Hospital, Harvard University, Boston, USA
| | | | | | | | - Ralph Santos Oliveira
- Brazilian Association of Radiopharmacy Brazil, Brazilian Nuclear Energy Commission - Nuclear Engineering Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Tamer Sakr
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Sarah Spreckelmeyer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
28
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
29
|
Yu X, Yang X, Nie H, Jiang W, He X, Ou C. Immunological role and prognostic value of somatostatin receptor family members in colon adenocarcinoma. Front Pharmacol 2023; 14:1255809. [PMID: 37900156 PMCID: PMC10603271 DOI: 10.3389/fphar.2023.1255809] [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: 07/09/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
Colon adenocarcinoma (COAD) is among the most prevalent cancers worldwide, ranking as the third most prevalent malignancy in incidence and mortality. The somatostatin receptor (SSTR) family comprises G-protein-coupled receptors (GPCRs), which couple to inhibitory G proteins (Gi and Go) upon binding to somatostatin (SST) analogs. GPCRs are involved in hormone release, neurotransmission, cell growth inhibition, and cancer suppression. However, their roles in COAD remain unclear. This study used bioinformatics to investigate the expression, prognosis, gene alterations, functional enrichment, and immunoregulatory effects of the SSTR family members in COAD. SSTR1-4 are differentially downregulated in COAD, and low SSTR2 expression indicates poor survival. Biological processes and gene expression enrichment of the SSTR family in COAD were further analyzed using the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology. A strong correlation was observed between SSTR expression and immune cell infiltration. We also quantified SSTR2 expression in 25 COAD samples and adjacent normal tissues using quantitative real-time polymerase chain reaction. We analyzed its correlation with the dendritic cell-integrin subunit alpha X marker gene. The biomarker exploration of the solid tumors portal was used to confirm the correlation between SSTR2 with immunomodulators and immunotherapy responses. Our results identify SSTR2 as a promising target for COAD immunotherapy. Our findings provide new insights into the biological functions of the SSTR family and their implications for the prognosis of COAD.
Collapse
Affiliation(s)
- Xiaoqian Yu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuejie Yang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Nie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenying Jiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Kersting D, Sandach P, Sraieb M, Wiesweg M, Metzenmacher M, Darwiche K, Oezkan F, Bölükbas S, Stuschke M, Umutlu L, Nader M, Hamacher R, Fendler WP, Wienker J, Eberhardt WEE, Schuler M, Herrmann K, Hautzel H. 68Ga-SSO-120 PET for Initial Staging of Small Cell Lung Cancer Patients: A Single-Center Retrospective Study. J Nucl Med 2023; 64:1540-1549. [PMID: 37474272 DOI: 10.2967/jnumed.123.265664] [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: 03/06/2023] [Revised: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
PET imaging using the somatostatin receptor 2 (SSTR2) antagonist satoreotide trizoxetan (SSO-120, previously OPS-202) could offer accurate tumor detection and screening for SSTR2-antagonist radionuclide therapy in patients with SSTR2-expressing small cell lung cancer (SCLC). The aim of this single-center study was to investigate tumor uptake and detection rates of 68Ga-SSO-120 in comparison to 18F-FDG PET in the initial staging of SCLC patients. Methods: Patients with newly diagnosed SCLC who underwent additional whole-body 68Ga-SSO-120 PET/CT during the initial diagnostic workup were retrospectively included. The mean administered activity was 139 MBq, and the mean uptake time was 60 min. Gold-standard staging 18F-FDG PET/CT was evaluated if available within 2 wk before or after 68Ga-SSO-120 PET if morphologic differences in CT images were absent. 68Ga-SSO-120- or 18F-FDG-positive lesions were reported in 7 anatomic regions (primary tumor, thoracic lymph node metastases, and distant metastases including pleural, contralateral pulmonary, liver, bone, and other) according to the TNM classification for lung cancer (eighth edition). Consensus TNM staging (derived from CT, endobronchial ultrasound-guided transbronchial needle aspiration, PET, and brain MRI) by a clinical tumor board served as the reference standard. Results: Thirty-one patients were included, 12 with limited and 19 with extensive disease according to the Veterans Administration Lung Study Group classification. 68Ga-SSO-120-positive tumor was detected in all patients (100%) and in 90 of the 217 evaluated regions (41.5%). Thirteen patients (42.0%) had intense average 68Ga-SSO-120 uptake (region-based mean SUVmax ≥ 10); 28 patients (90.3%) had average 68Ga-SSO-120 uptake greater than liver uptake (region-based mean peak tumor-to-liver ratio > 1). In 25 patients with evaluable 18F-FDG PET, primary tumor, thoracic lymph node metastases, and distant metastases were detected in 100%, 92%, and 64%, respectively, of all investigated patients by 68Ga-SSO-120 and in 100%, 92%, and 56%, respectively, by 18F-FDG PET. 68Ga-SSO-120 PET detected additional contralateral lymph node, liver, and brain metastases in 1, 1, and 2 patients, respectively (no histopathology available), and 18F-FDG PET detected additional contralateral lymph node metastases in 3 patients (1 confirmed, 1 systematic endobronchial ultrasound-guided transbronchial needle aspiration-negative, and 1 without available histopathology). None of these differences altered Veterans Administration Lung Study Group staging. The region-based monotonic correlation between 68Ga-SSO-120 and 18F-FDG uptake was low (Spearman ρ = 0.26-0.33). Conclusion: 68Ga-SSO-120 PET offers high diagnostic precision with comparable detection rates and additional complementary information to the gold standard, 18F-FDG PET. Consistent uptake in most patients warrants exploration of SSTR2-directed radionuclide therapy.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Patrick Sandach
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Miriam Sraieb
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Marcel Wiesweg
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Metzenmacher
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kaid Darwiche
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Filiz Oezkan
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Servet Bölükbas
- Department of Thoracic Surgery and Thoracic Endoscopy, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lale Umutlu
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Michael Nader
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Johannes Wienker
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Wilfried E E Eberhardt
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Martin Schuler
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Hubertus Hautzel
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
31
|
Gabiache G, Zadro C, Rozenblum L, Vezzosi D, Mouly C, Thoulouzan M, Guimbaud R, Otal P, Dierickx L, Rousseau H, Trepanier C, Dercle L, Mokrane FZ. Image-Guided Precision Medicine in the Diagnosis and Treatment of Pheochromocytomas and Paragangliomas. Cancers (Basel) 2023; 15:4666. [PMID: 37760633 PMCID: PMC10526298 DOI: 10.3390/cancers15184666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
In this comprehensive review, we aimed to discuss the current state-of-the-art medical imaging for pheochromocytomas and paragangliomas (PPGLs) diagnosis and treatment. Despite major medical improvements, PPGLs, as with other neuroendocrine tumors (NETs), leave clinicians facing several challenges; their inherent particularities and their diagnosis and treatment pose several challenges for clinicians due to their inherent complexity, and they require management by multidisciplinary teams. The conventional concepts of medical imaging are currently undergoing a paradigm shift, thanks to developments in radiomic and metabolic imaging. However, despite active research, clinical relevance of these new parameters remains unclear, and further multicentric studies are needed in order to validate and increase widespread use and integration in clinical routine. Use of AI in PPGLs may detect changes in tumor phenotype that precede classical medical imaging biomarkers, such as shape, texture, and size. Since PPGLs are rare, slow-growing, and heterogeneous, multicentric collaboration will be necessary to have enough data in order to develop new PPGL biomarkers. In this nonsystematic review, our aim is to present an exhaustive pedagogical tool based on real-world cases, dedicated to physicians dealing with PPGLs, augmented by perspectives of artificial intelligence and big data.
Collapse
Affiliation(s)
- Gildas Gabiache
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Charline Zadro
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Laura Rozenblum
- Department of Nuclear Medicine, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 75013 Paris, France
| | - Delphine Vezzosi
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | - Céline Mouly
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | | | - Rosine Guimbaud
- Department of Oncology, Rangueil University Hospital, 31400 Toulouse, France
| | - Philippe Otal
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Lawrence Dierickx
- Department of Nuclear Medicine, IUCT-Oncopole, 31059 Toulouse, France;
| | - Hervé Rousseau
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Christopher Trepanier
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Laurent Dercle
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fatima-Zohra Mokrane
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| |
Collapse
|
32
|
Şen F, Sheikh GT, von Hinten J, Schindele A, Kircher M, Dierks A, Pfob CH, Serfling SE, Buck AK, Pelzer T, Higuchi T, Weich A, Bundschuh RA, Werner RA, Lapa C. In-Vivo Somatostatin-Receptor Expression in Small Cell Lung Cancer as a Prognostic Image Biomarker and Therapeutic Target. Cancers (Basel) 2023; 15:3595. [PMID: 37509258 PMCID: PMC10377032 DOI: 10.3390/cancers15143595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Given the dismal prognosis of small cell lung cancer (SCLC), novel therapeutic targets are urgently needed. We aimed to evaluate whether SSTR expression, as assessed by positron emission tomography (PET), can be applied as a prognostic image biomarker and determined subjects eligible for peptide receptor radionuclide therapy (PRRT). METHODS A total of 67 patients (26 females; age, 41-80 years) with advanced SCLC underwent SSTR-directed PET/computed tomography (somatostatin receptor imaging, SRI). SRI-avid tumor burden was quantified by maximum standardized uptake values (SUVmax) and tumor-to-liver ratios (T/L) of the most intense SCLC lesion. Scan findings were correlated with progression-free (PFS) and overall survival (OS). In addition, subjects eligible for SSTR-directed radioligand therapy were identified, and treatment outcome and toxicity profile were recorded. RESULTS On a patient basis, 36/67 (53.7%) subjects presented with mainly SSTR-positive SCLC lesions (>50% lesions positive); in 10/67 patients (14.9%), all lesions were positive. The median SUVmax was found to be 8.5, while the median T/L was 1.12. SRI-uptake was not associated with PFS or OS, respectively (SUVmax vs. PFS, ρ = 0.13 with p = 0.30 and vs. OS, ρ = 0.00 with p = 0.97; T/L vs. PFS, ρ = 0.07 with p = 0.58 and vs. OS, ρ = -0.05 with p = 0.70). PRRT was performed in 14 patients. One patient succumbed to treatment-independent infectious complications immediately after PRRT. In the remaining 13 subjects, disease control was achieved in 5/13 (38.5%) with a single patient achieving a partial response (stable disease in the remainder). In the sub-group of responding patients, PFS and OS were 357 days and 480 days, respectively. CONCLUSIONS SSTR expression as detected by SRI is not predictive of outcome in patients with advanced SCLC. However, it might serve as a therapeutic target in selected patients.
Collapse
Affiliation(s)
- Feyza Şen
- Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Würzburg, Germany
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
- Department of Nuclear Medicine, Pendik Training and Research Hospital, Marmara University, 34722 İstanbul, Turkey
| | - Gabriel T Sheikh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Johannes von Hinten
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Andreas Schindele
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Christian H Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Sebastian E Serfling
- Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Würzburg, Germany
| | - Theo Pelzer
- Department of Internal Medicine I, Pulmonology, University Hospital Wuerzburg, 97080 Würzburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Würzburg, Germany
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Alexander Weich
- Department of Internal Medicine II, Gastroenterology, University Hospital Wuerzburg, 97080 Würzburg, Germany
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Würzburg, Germany
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, 86154 Augsburg, Germany
| |
Collapse
|
33
|
Burkett BJ, Bartlett DJ, McGarrah PW, Lewis AR, Johnson DR, Berberoğlu K, Pandey MK, Packard AT, Halfdanarson TR, Hruska CB, Johnson GB, Kendi AT. A Review of Theranostics: Perspectives on Emerging Approaches and Clinical Advancements. Radiol Imaging Cancer 2023; 5:e220157. [PMID: 37477566 PMCID: PMC10413300 DOI: 10.1148/rycan.220157] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 07/22/2023]
Abstract
Theranostics is the combination of two approaches-diagnostics and therapeutics-applied for decades in cancer imaging using radiopharmaceuticals or paired radiopharmaceuticals to image and selectively treat various cancers. The clinical use of theranostics has increased in recent years, with U.S. Food and Drug Administration (FDA) approval of lutetium 177 (177Lu) tetraazacyclododecane tetraacetic acid octreotate (DOTATATE) and 177Lu-prostate-specific membrane antigen vector-based radionuclide therapies. The field of theranostics has imminent potential for emerging clinical applications. This article reviews critical areas of active clinical advancement in theranostics, including forthcoming clinical trials advancing FDA-approved and emerging radiopharmaceuticals, approaches to dosimetry calculations, imaging of different radionuclide therapies, expanded indications for currently used theranostic agents to treat a broader array of cancers, and emerging ideas in the field. Keywords: Molecular Imaging, Molecular Imaging-Cancer, Molecular Imaging-Clinical Translation, Molecular Imaging-Target Development, PET/CT, SPECT/CT, Radionuclide Therapy, Dosimetry, Oncology, Radiobiology © RSNA, 2023.
Collapse
Affiliation(s)
- Brian J. Burkett
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - David J. Bartlett
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Patrick W. McGarrah
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Akeem R. Lewis
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Derek R. Johnson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Kezban Berberoğlu
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Mukesh K. Pandey
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Annie T. Packard
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Thorvardur R. Halfdanarson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Carrie B. Hruska
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Geoffrey B. Johnson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - A. Tuba Kendi
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| |
Collapse
|
34
|
Handula M, Beekman S, Konijnenberg M, Stuurman D, de Ridder C, Bruchertseifer F, Morgenstern A, Denkova A, de Blois E, Seimbille Y. First preclinical evaluation of [ 225Ac]Ac-DOTA-JR11 and comparison with [ 177Lu]Lu-DOTA-JR11, alpha versus beta radionuclide therapy of NETs. EJNMMI Radiopharm Chem 2023; 8:13. [PMID: 37389800 DOI: 10.1186/s41181-023-00197-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND The [177Lu]Lu-DOTA-TATE mediated peptide receptor radionuclide therapy (PRRT) of neuroendocrine tumors (NETs) is sometimes leading to treatment resistance and disease recurrence. An interesting alternative could be the somatostatin antagonist, [177Lu]Lu-DOTA-JR11, that demonstrated better biodistribution profile and higher tumor uptake than [177Lu]Lu-DOTA-TATE. Furthermore, treatment with alpha emitters showed improvement of the therapeutic index of PRRT due to the high LET offered by the alpha particles compared to beta emitters. Therefore, [225Ac]Ac-DOTA-JR11 can be a potential candidate to improve the treatment of NETs (Graphical abstract). DOTA-JR11 was radiolabeled with [225Ac]Ac(NO3)3 and [177Lu]LuCl3. Stability studies were performed in phosphate buffered saline (PBS) and mouse serum. In vitro competitive binding assay has been carried out in U2OS-SSTR2 + cells for natLa-DOTA-JR11, natLu-DOTA-JR11 and DOTA-JR11. Ex vivo biodistribution studies were performed in mice inoculated with H69 cells at 4, 24, 48 and 72 h after injection of [225Ac]Ac-DOTA-JR11. A blocking group was included to verify uptake specificity. Dosimetry of selected organs was determined for [225Ac]Ac-DOTA-JR11 and [177Lu]Lu-DOTA-JR11. RESULTS [225Ac]Ac-DOTA-JR11 has been successfully prepared and obtained in high radiochemical yield (RCY; 95%) and radiochemical purity (RCP; 94%). [225Ac]Ac-DOTA-JR11 showed reasonably good stability in PBS (77% intact radiopeptide at 24 h after incubation) and in mouse serum (~ 81% intact radiopeptide 24 h after incubation). [177Lu]Lu-DOTA-JR11 demonstrated excellent stability in both media (> 93%) up to 24 h post incubation. Competitive binding assay revealed that complexation of DOTA-JR11 with natLa and natLu did not affect its binding affinity to SSTR2. Similar biodistribution profiles were observed for both radiopeptides, however, higher uptake was noticed in the kidneys, liver and bone for [225Ac]Ac-DOTA-JR11 than [177Lu]Lu-DOTA-JR11. CONCLUSION [225Ac]Ac-DOTA-JR11 showed a higher absorbed dose in the kidneys compared to [177Lu]Lu-DOTA-JR11, which may limit further studies with this radiopeptide. However, several strategies can be explored to reduce nephrotoxicity and offer opportunities for future clinical investigations with [225Ac]Ac-DOTA-JR11.
Collapse
Affiliation(s)
- Maryana Handula
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Department of Experimental Urology, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Department of Experimental Urology, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | | | | | - Antonia Denkova
- Applied Radiation and Isotopes, Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD, Rotterdam, The Netherlands.
- Life Sciences Division, TRIUMF, Vancouver, BC, V6T 2A3, Canada.
| |
Collapse
|
35
|
Adnan A, Basu S. Somatostatin Receptor Targeted PET-CT and Its Role in the Management and Theranostics of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics (Basel) 2023; 13:2154. [PMID: 37443548 DOI: 10.3390/diagnostics13132154] [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: 05/01/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Somatostatin receptor (SSTR) agonist-based Positron Emission Tomography-Computed Tomography (PET-CT) imaging is nowadays the mainstay for the assessment and diagnostic imaging of neuroendocrine neoplasms (NEN), especially in well-differentiated neuroendocrine tumors (NET) (World Health Organization (WHO) grade I and II). Major clinical indications for SSTR imaging are primary staging and metastatic workup, especially (a) before surgery, (b) detection of unknown primary in metastatic NET, (c) patient selection for theranostics and appropriate therapy, especially peptide receptor radionuclide therapy (PRRT), while less major indications include treatment response evaluation on and disease prognostication. Dual tracer PET-CT imaging using SSTR targeted PET tracers, viz. [68Ga]Ga-DOTA-Tyr3-Octreotate (DOTA-TATE) and [68Ga]Ga-DOTA-NaI3-Octreotide (DOTA-NOC), and fluorodeoxyglucose (FDG), have recently gained widespread acceptance for better assessment of whole-body tumor biology compared to single-site histopathology, in terms of being non-invasive and the ability to assess inter- and intra-tumoral heterogeneity on a global scale. FDG uptake has been identified as independent adverse risk factor in various studies. Recently, somatostatin receptor antagonists have been shown to be more sensitive and specific in detecting the disease. The aim of this review article is to summarize the clinical importance of SSTR-based imaging in the clinical management of neuroendocrine and related tumors.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
| |
Collapse
|
36
|
Lepareur N, Ramée B, Mougin-Degraef M, Bourgeois M. Clinical Advances and Perspectives in Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1733. [PMID: 37376181 DOI: 10.3390/pharmaceutics15061733] [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: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted radionuclide therapy has become increasingly prominent as a nuclear medicine subspecialty. For many decades, treatment with radionuclides has been mainly restricted to the use of iodine-131 in thyroid disorders. Currently, radiopharmaceuticals, consisting of a radionuclide coupled to a vector that binds to a desired biological target with high specificity, are being developed. The objective is to be as selective as possible at the tumor level, while limiting the dose received at the healthy tissue level. In recent years, a better understanding of molecular mechanisms of cancer, as well as the appearance of innovative targeting agents (antibodies, peptides, and small molecules) and the availability of new radioisotopes, have enabled considerable advances in the field of vectorized internal radiotherapy with a better therapeutic efficacy, radiation safety and personalized treatments. For instance, targeting the tumor microenvironment, instead of the cancer cells, now appears particularly attractive. Several radiopharmaceuticals for therapeutic targeting have shown clinical value in several types of tumors and have been or will soon be approved and authorized for clinical use. Following their clinical and commercial success, research in that domain is particularly growing, with the clinical pipeline appearing as a promising target. This review aims to provide an overview of current research on targeting radionuclide therapy.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, 35000 Rennes, France
- Inserm, INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)-UMR 1317, Univ Rennes, 35000 Rennes, France
| | - Barthélémy Ramée
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
| | - Marie Mougin-Degraef
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
- Groupement d'Intérêt Public ARRONAX, 1 Rue Aronnax, 44817 Saint Herblain, France
| |
Collapse
|
37
|
Merola E, Grana CM. Peptide Receptor Radionuclide Therapy (PRRT): Innovations and Improvements. Cancers (Basel) 2023; 15:2975. [PMID: 37296936 PMCID: PMC10251822 DOI: 10.3390/cancers15112975] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are tumors originating from neuroendocrine cells distributed throughout the human body. With an increasing incidence over the past few decades, they represent a highly heterogeneous group of neoplasms, mostly expressing somatostatin receptors (SSTRs) on their cell surface. Peptide receptor radionuclide therapy (PRRT) has emerged as a crucial strategy for treating advanced, unresectable neuroendocrine tumors by administering radiolabeled somatostatin analogs intravenously to target SSTRs. This article will focus on the multidisciplinary theranostic approach, treatment effectiveness (such as response rates and symptom relief), patient outcomes, and toxicity profile of PRRT for NEN patients. We will review the most significant studies, such as the phase III NETTER-1 trial, and discuss promising new radiopharmaceuticals, including alpha-emitting radionuclide-labeled somatostatin analogs and SSTR antagonists.
Collapse
Affiliation(s)
- Elettra Merola
- Gastroenterology Unit, G.B. Grassi Hospital (ASL Roma 3), Lido di Ostia, 00122 Rome, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IRCCS European Institute of Oncology, 20141 Milan, Italy;
| |
Collapse
|
38
|
Abstract
Neuroendocrine neoplasms (NENs) are tumors originating from neuroendocrine cells distributed throughout the human body. With an increasing incidence over the past few decades, they represent a highly heterogeneous group of neoplasms, mostly expressing somatostatin receptors (SSTRs) on their cell surface. Peptide receptor radionuclide therapy (PRRT) has emerged as a crucial strategy for treating advanced, unresectable neuroendocrine tumors by administering radiolabeled somatostatin analogs intravenously to target SSTRs. This article will focus on the multidisciplinary theranostic approach, treatment effectiveness (such as response rates and symptom relief), patient outcomes, and toxicity profile of PRRT for NEN patients. We will review the most significant studies, such as the phase III NETTER-1 trial, and discuss promising new radiopharmaceuticals, including alpha-emitting radionuclide-labeled somatostatin analogs and SSTR antagonists.
Collapse
Affiliation(s)
- Elettra Merola
- Gastroenterology Unit, G.B. Grassi Hospital (ASL Roma 3), Lido di Ostia, 00122 Rome, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IRCCS European Institute of Oncology, 20141 Milan, Italy
| |
Collapse
|
39
|
Imperiale A, Jha A, Meuter L, Nicolas GP, Taïeb D, Pacak K. The Emergence of Somatostatin Antagonist-Based Theranostics: Paving the Road Toward Another Success? J Nucl Med 2023; 64:682-684. [PMID: 36759198 PMCID: PMC10152128 DOI: 10.2967/jnumed.123.265406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Affiliation(s)
- Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, ICANS, Strasbourg University, Strasbourg, France;
- Molecular Imaging-DRHIM, IPHC, UMR-7178, CNRS/Unistra, Strasbourg, France
| | - Abhishek Jha
- Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, Maryland
| | - Leah Meuter
- Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, Maryland
| | - Guillaume P Nicolas
- Division of Nuclear Medicine, Center for Neuroendocrine and Endocrine Tumors, University Hospital Basel, Basel, Switzerland; and
| | - David Taïeb
- La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Nock BA, Kanellopoulos P, Joosten L, Mansi R, Maina T. Peptide Radioligands in Cancer Theranostics: Agonists and Antagonists. Pharmaceuticals (Basel) 2023; 16:ph16050674. [PMID: 37242457 DOI: 10.3390/ph16050674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical success of radiolabeled somatostatin analogs in the diagnosis and therapy-"theranostics"-of tumors expressing the somatostatin subtype 2 receptor (SST2R) has paved the way for the development of a broader panel of peptide radioligands targeting different human tumors. This approach relies on the overexpression of other receptor-targets in different cancer types. In recent years, a shift in paradigm from internalizing agonists to antagonists has occurred. Thus, SST2R-antagonist radioligands were first shown to accumulate more efficiently in tumor lesions and clear faster from the background in animal models and patients. The switch to receptor antagonists was soon adopted in the field of radiolabeled bombesin (BBN). Unlike the stable cyclic octapeptides used in the case of somatostatin, BBN-like peptides are linear, fast to biodegradable and elicit adverse effects in the body. Thus, the advent of BBN-like antagonists provided an elegant way to obtain effective and safe radiotheranostics. Likewise, the pursuit of gastrin and exendin antagonist-based radioligands is advancing with exciting new outcomes on the horizon. In the present review, we discuss these developments with a focus on clinical results, commenting on challenges and opportunities for personalized treatment of cancer patients by means of state-of-the-art antagonist-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Berthold A Nock
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| | | | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| |
Collapse
|
41
|
Gervasoni S, Guccione C, Fanti V, Bosin A, Cappellini G, Golosio B, Ruggerone P, Malloci G. Molecular simulations of SSTR2 dynamics and interaction with ligands. Sci Rep 2023; 13:4768. [PMID: 36959237 PMCID: PMC10036620 DOI: 10.1038/s41598-023-31823-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
The cyclic peptide hormone somatostatin regulates physiological processes involved in growth and metabolism, through its binding to G-protein coupled somatostatin receptors. The isoform 2 (SSTR2) is of particular relevance for the therapy of neuroendocrine tumours for which different analogues to somatostatin are currently in clinical use. We present an extensive and systematic computational study on the dynamics of SSTR2 in three different states: active agonist-bound, inactive antagonist-bound and apo inactive. We exploited the recent burst of SSTR2 experimental structures to perform μs-long multi-copy molecular dynamics simulations to sample conformational changes of the receptor and rationalize its binding to different ligands (the agonists somatostatin and octreotide, and the antagonist CYN154806). Our findings suggest that the apo form is more flexible compared to the holo ones, and confirm that the extracellular loop 2 closes upon the agonist octreotide but not upon the antagonist CYN154806. Based on interaction fingerprint analyses and free energy calculations, we found that all peptides similarly interact with residues buried into the binding pocket. Conversely, specific patterns of interactions are found with residues located in the external portion of the pocket, at the basis of the extracellular loops, particularly distinguishing the agonists from the antagonist. This study will help in the design of new somatostatin-based compounds for theranostics of neuroendocrine tumours.
Collapse
Affiliation(s)
- Silvia Gervasoni
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Camilla Guccione
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Viviana Fanti
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
- Istituto Nazionale di Fisica Nucleare, Sezione di Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Andrea Bosin
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Giancarlo Cappellini
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Bruno Golosio
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
- Istituto Nazionale di Fisica Nucleare, Sezione di Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, 09042, Monserrato (Cagliari), Italy.
| |
Collapse
|
42
|
Selective targeting of gold nanoparticles for radiosensitization of somatostatin 2 receptor-expressing cancer cells. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
|
43
|
Ahenkorah S, Cawthorne C, Murce E, Deroose CM, Cardinaels T, Seimbille Y, Bormans G, Ooms M, Cleeren F. Direct comparison of [18F]AlF-NOTA-JR11 and [18F]AlF-NOTA-octreotide for PET imaging of neuroendocrine tumors: Antagonist versus agonist. Nucl Med Biol 2023; 118-119:108338. [PMID: 37018875 DOI: 10.1016/j.nucmedbio.2023.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND [18F]AlF-NOTA-octreotide is an 18F-labeled somatostatin analogue which is a good clinical alternative for 68Ga-labeled somatostatin analogues. However, radiolabeled somatostatin receptor (SSTR) antagonists might outperform agonists regarding imaging sensitivity of neuroendocrine tumors (NETs). No direct comparison between the antagonist [18F]AlF-NOTA-JR11 and the agonist [18F]AlF-NOTA-octreotide as SSTR PET probes is available. Herein, we present the radiosynthesis of [18F]AlF-NOTA-JR11 and compare its NETs imaging properties directly with the established agonist radioligand [18F]AlF-NOTA-octreotide preclinically. METHODS [18F]AlF-NOTA-JR11 was synthesized in an automated synthesis module. The in vitro binding characteristics (IC50) of [natF]AlF-NOTA-JR11 and [natF]AlF-NOTA-octreotide were evaluated and the in vitro stability of [18F]AlF-NOTA-JR11 was determined in human serum. In vitro cell binding and internalization was performed with [18F]AlF-NOTA-JR11 and [18F]AlF-NOTA-octreotide using SSTR2 expressing cells and the pharmacokinetics were evaluated using μPET/CT in mice bearing BON1.SSTR2 tumor xenografts. RESULTS Excellent binding affinity for SSTR2 was found for [natF]AlF-NOTA-octreotide (IC50 of 25.7 ± 7.9 nM). However, the IC50 value for [natF]AlF-NOTA-JR11 (290.6 ± 71 nM) was 11-fold higher compared to [natF]AlF-NOTA-octreotide, indicating lower affinity for SSTR2. [18F]AlF-NOTA-JR11 was obtained in a good RCY (50 ± 6 %) but with moderate RCP of 94 ± 1 %. [18F]AlF-NOTA-JR11 demonstrated excellent stability in human serum (>95 % after 240 min). 2.7-fold higher cell binding was observed for [18F]AlF-NOTA-JR11 as compared to [18F]AlF-NOTA-octreotide after 60 min. μPET/CT images demonstrated comparable pharmacokinetics and tumor uptake between [18F]AlF-NOTA-JR11 (SUVmax: 3.7 ± 0.8) and [18F]AlF-NOTA-octreotide (SUVmax: 3.6 ± 0.4). CONCLUSIONS [18F]AlF-NOTA-JR11 was obtained in good RCY, albeit with a moderate RCP. The cell binding study showed significant higher binding of [18F]AlF-NOTA-JR11 compared to [18F]AlF-NOTA-octreotide, despite the higher IC50 value of AlF-NOTA-JR11. However, pharmacokinetics and in vivo tumor uptake was comparable for both radiotracers. Novel Al18F-labeled derivatives of JR11 with higher SSTR2 affinity should be developed for increased tumor uptake and NET imaging sensitivity.
Collapse
|
44
|
Makris G, Li Y, Gallazzi F, Kuchuk M, Wang J, Lewis MR, Jurisson SS, Hennkens HM. Evaluation of Re/ 99mTc-labeled somatostatin receptor-targeting peptide complexes synthesized via direct metal cyclization. RADIOCHIM ACTA 2023. [DOI: 10.1515/ract-2022-0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
With interest in the development of somatostatin receptor (SSTR) targeting agents for potential application in diagnostic SPECT imaging (99mTc) or Peptide Radionuclide Receptor Therapy (PRRT, 186Re or 188Re) of neuroendocrine tumors, we present herein 99mTc/Re (radio)complexes synthesized by the integrated (radio)labeling approach of peptide cyclization via metal complexation. In particular, we utilized the potent SSTR2 peptide antagonist sequence DOTA-4-NO2-Phe-c(DCys-Tyr-DTrp-Lys-Thr-Cys)-DTyr-NH2 (DOTA-sst2-ANT) and report the syntheses and in vitro evaluations of its respective [99mTc]Tc/Re-cyclized peptides ([99mTc]Tc/Re-cyc-DOTA-sst2-ANT). The Re-cyc-DOTA-sst2-ANT complex was synthesized via an on-resin Re(V)-cyclization reaction using the ReOCl3(PPh3)2 precursor and consisted of three isomers characterized by LC–ESI-MS. The [99mTc]Tc-cyclized analogue was prepared via a ligand exchange reaction of the [99mTc][TcO]3+ core through a [99mTc]Tc-glucoheptonate intermediate with linear DOTA-sst2-ANT and was characterized by comparative HPLC studies against Re-cyc-DOTA-sst2-ANT. Good in vitro binding affinity was demonstrated in SSTR-expressing cells (AR42J) by the Re-cyc-DOTA-sst2-ANT major isomer, similar to the potent binder Lu-DOTA-sst2-ANT, in which the Lu metal was complexed by the bifunctional chelator DOTA versus via peptide cyclization. [99mTc]Tc-cyc-DOTA-sst2-ANT was obtained in high radiochemical yield, also with an elution pattern of three isomers observed by HPLC analysis, which were comparable yet not identical to those of Re-cyc-DOTA-sst2-ANT. The [99mTc]Tc-tracer complex was shown to be hydrophilic, and stability studies at 4 h demonstrated that it remained intact in both PBS and in rat serum, with low non-specific rat serum protein binding, while exhibiting more moderate stability in 1 mM cysteine. These findings demonstrate that direct Re/[99mTc]Tc-cyclization of DOTA-sst2-ANT is feasible and may be used as an alternative approach to the bifunctional chelate labeling strategy. However, given that the non-radioactive (Re) and radiotracer (99mTc) analogues are not identical and both form isomeric products in equilibrium, additional design modifications will be necessary prior to in vivo application of [99mTc]Tc/Re-cyc-DOTA-sst2-ANT.
Collapse
Affiliation(s)
- George Makris
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
| | - Yawen Li
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| | - Fabio Gallazzi
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
- Molecular Interactions Core, University of Missouri , Columbia , MO 65211 , USA
| | - Marina Kuchuk
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
| | - Jing Wang
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
- Institute of Nuclear Physics and Chemistry, China Academy of Engineering Physics , Mianyang , Sichuan 621900 , P. R. China
| | - Michael R. Lewis
- Department of Veterinary Medicine and Surgery , University of Missouri , Columbia , MO 65211 , USA
- Research Service, Harry S. Truman Memorial Veterans’ Hospital , Columbia , MO 65201 , USA
| | - Silvia S. Jurisson
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| | - Heather M. Hennkens
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| |
Collapse
|
45
|
Pang XX, Xie L, Yao WJ, Liu XX, Pan B, Chen N. Advancements of molecular imaging and radiomics in pancreatic carcinoma. World J Radiol 2023; 15:10-19. [PMID: 36721672 PMCID: PMC9884334 DOI: 10.4329/wjr.v15.i1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Despite the recent progress of medical technology in the diagnosis and treatment of tumors, pancreatic carcinoma remains one of the most malignant tumors, with extremely poor prognosis partly due to the difficulty in early and accurate imaging evaluation. This paper focuses on the research progress of magnetic resonance imaging, nuclear medicine molecular imaging and radiomics in the diagnosis of pancreatic carcinoma. We also briefly described the achievements of our team in this field, to facilitate future research and explore new technologies to optimize diagnosis of pancreatic carcinoma.
Collapse
Affiliation(s)
- Xiao-Xi Pang
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Liang Xie
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Wen-Jun Yao
- Department of Radiology, The Second affiliated hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Xiu-Xia Liu
- Department of Nuclear Medicine, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui Province, China
| | - Bo Pan
- PET/CT Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Ni Chen
- Department of Nuclear Medicine, School of Basic Medicine Anhui Medical University, Hefei 230032, Anhui Province, China
| |
Collapse
|
46
|
Csaba Z, Dournaud P. Internalization of somatostatin receptors in brain and periphery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:43-57. [PMID: 36813365 DOI: 10.1016/bs.pmbts.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Somatostatin (SRIF) is a neuropeptide that acts as an important regulator of both endocrine and exocrine secretion and modulates neurotransmission in the central nervous system (CNS). SRIF also regulates cell proliferation in normal tissues and tumors. The physiological actions of SRIF are mediated by a family of five G protein-coupled receptors, called somatostatin receptor (SST) SST1, SST2, SST3, SST4, SST5. These five receptors share similar molecular structure and signaling pathways but they display marked differences in their anatomical distribution, subcellular localization and intracellular trafficking. The SST subtypes are widely distributed in the CNS and peripheral nervous system, in many endocrine glands and tumors, particularly of neuroendocrine origin. In this review, we focus on the agonist-dependent internalization and recycling of the different SST subtypes in vivo in the CNS, peripheral organs and tumors. We also discuss the physiological, pathophysiological and potential therapeutic effects of the intracellular trafficking of SST subtypes.
Collapse
Affiliation(s)
- Zsolt Csaba
- Université Paris Cité, NeuroDiderot, Inserm UMR, Paris, France
| | - Pascal Dournaud
- Université Paris Cité, NeuroDiderot, Inserm UMR, Paris, France.
| |
Collapse
|
47
|
Shi M, Jakobsson V, Greifenstein L, Khong PL, Chen X, Baum RP, Zhang J. Alpha-peptide receptor radionuclide therapy using actinium-225 labeled somatostatin receptor agonists and antagonists. Front Med (Lausanne) 2022; 9:1034315. [PMID: 36569154 PMCID: PMC9767967 DOI: 10.3389/fmed.2022.1034315] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has over the last two decades emerged as a very promising approach to treat neuroendocrine tumors (NETs) with rapidly expanding clinical applications. By chelating a radiometal to a somatostatin receptor (SSTR) ligand, radiation can be delivered to cancer cells with high precision. Unlike conventional external beam radiotherapy, PRRT utilizes primarily β or α radiation derived from nuclear decay, which causes damage to cancer cells in the immediate proximity by irreversible direct or indirect ionization of the cells' DNA, which induces apoptosis. In addition, to avoid damage to surrounding normal cells, PRRT privileges the use of radionuclides that have little penetrating and more energetic (and thus more ionizing) radiations. To date, the most frequently radioisotopes are β- emitters, particularly Yttrium-90 (90Y) and Lutetium-177 (177Lu), labeled SSTR agonists. Current development of SSTR-targeting is triggering the shift from using SSTR agonists to antagonists for PRRT. Furthermore, targeted α-particle therapy (TAT), has attracted special attention for the treatment of tumors and offers an improved therapeutic option for patients resistant to conventional treatments or even beta-irradiation treatment. Due to its short range and high linear energy transfer (LET), α-particles significantly damage the targeted cancer cells while causing minimal cytotoxicity toward surrounding normal tissue. Actinium-225 (225Ac) has been developed into potent targeting drug constructs including somatostatin-receptor-based radiopharmaceuticals and is in early clinical use against multiple neuroendocrine tumor types. In this article, we give a review of preclinical and clinical applications of 225Ac-PRRT in NETs, discuss the strengths and challenges of 225Ac complexes being used in PRRT; and envision the prospect of 225Ac-PRRT as a future alternative in the treatment of NETs.
Collapse
Affiliation(s)
- Mengqi Shi
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Academy for Precision Oncology, International Centers for Precision Oncology (ICPO), Wiesbaden, Germany
| | - Lukas Greifenstein
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| | - Pek-Lan Khong
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Agency for Science, Technology, and Research (A*STAR), Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
48
|
Rajamohan N, Khasawneh H, Singh A, Suman G, Johnson GB, Majumder S, Halfdanarson TR, Goenka AH. PET/CT and PET/MRI in neuroendocrine neoplasms. Abdom Radiol (NY) 2022; 47:4058-4072. [PMID: 35426497 DOI: 10.1007/s00261-022-03516-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 01/18/2023]
Abstract
Advanced molecular imaging has come to play an integral role in the management of gastro-entero-pancreatic neuroendocrine neoplasms (GEP-NENs). Somatostatin receptor (SSTR) PET has now emerged as the reference standard for the evaluation of NENs and is particularly critical in the context of peptide receptor radionuclide therapy (PRRT) eligibility. SSTR PET/MRI with liver-specific contrast agent has a strong potential for one-stop-shop multiparametric evaluation of GEP-NENs. 18F-FDG is a complementary radiotracer to SSTR, especially in the context of high-grade neuroendocrine neoplasms. Knowledge gaps in quantitative evaluation of molecular imaging studies and their role in assessment of response to PRRT and combination therapies are active research areas. Novel radiotracers have the potential to overcome existing limitations in the molecular imaging of GEP-NENs. The purpose of this article is to provide an overview of the current trends, pitfalls, and recent advancements of molecular imaging for GEP-NENs.
Collapse
Affiliation(s)
- Naveen Rajamohan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Aparna Singh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA.
| |
Collapse
|
49
|
Hofland J, Brabander T, Verburg FA, Feelders RA, de Herder WW. Peptide Receptor Radionuclide Therapy. J Clin Endocrinol Metab 2022; 107:3199-3208. [PMID: 36198028 PMCID: PMC9693835 DOI: 10.1210/clinem/dgac574] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Indexed: 11/19/2022]
Abstract
The concept of using a targeting molecule labeled with a diagnostic radionuclide for using positron emission tomography or single photon emission computed tomography imaging with the potential to demonstrate that tumoricidal radiation can be delivered to tumoral sites by administration of the same or a similar targeting molecule labeled with a therapeutic radionuclide termed "theranostics." Peptide receptor radionuclide therapy (PRRT) with radiolabeled somatostatin analogs (SSAs) is a well-established second/third-line theranostic treatment for somatostatin receptor-positive well-differentiated (neuro-)endocrine neoplasms (NENs). PRRT with 177Lu-DOTATATE was approved by the regulatory authorities in 2017 and 2018 for selected patients with low-grade well-differentiated gastroenteropancreatic (GEP) NENs. It improves progression-free survival as well as quality of life of GEP NEN patients. Favorable symptomatic and biochemical responses using PRRT with 177Lu-DOTATATE have also been reported in patients with functioning metastatic GEP NENs like metastatic insulinomas, Verner Morrison syndromes (VIPomas), glucagonomas, and gastrinomas and patients with carcinoid syndrome. This therapy might also become a valuable therapeutic option for inoperable low-grade bronchopulmonary NENs, inoperable or progressive pheochromocytomas and paragangliomas, and medullary thyroid carcinomas. First-line PRRT with 177Lu-DOTATATE and combinations of this therapy with cytotoxic drugs are currently under investigation. New radiolabeled somatostatin receptor ligands include SSAs coupled with alpha radiation emitting radionuclides and somatostatin receptor antagonists coupled with radionuclides.
Collapse
Affiliation(s)
- Johannes Hofland
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC and Erasmus Cancer Institute, Rotterdam, The Netherlands
| | - Tessa Brabander
- Department of Radiology & Nuclear Medicine, ENETS Center of Excellence, Erasmus MC and Erasmus Cancer Institute, Rotterdam, The Netherlands
| | - Frederik A Verburg
- Department of Radiology & Nuclear Medicine, ENETS Center of Excellence, Erasmus MC and Erasmus Cancer Institute, Rotterdam, The Netherlands
| | - Richard A Feelders
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC and Erasmus Cancer Institute, Rotterdam, The Netherlands
| | - Wouter W de Herder
- Correspondence: Wouter W. de Herder, MD, PhD, Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC and Erasmus Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
50
|
Kanellopoulos P, Nock BA, Greifenstein L, Baum RP, Roesch F, Maina T. [ 68Ga]Ga-DATA 5m-LM4, a PET Radiotracer in the Diagnosis of SST 2R-Positive Tumors: Preclinical and First Clinical Results. Int J Mol Sci 2022; 23:ijms232314590. [PMID: 36498918 PMCID: PMC9740503 DOI: 10.3390/ijms232314590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Radiolabeled somatostatin subtype 2 receptor (SST2R)-antagonists have shown advantageous profiles for cancer theranostics compared with agonists. On the other hand, the newly introduced hybrid chelator (6-pentanoic acid)-6-(amino)methyl-1,4-diazepinetriacetate (DATA5m) rapidly binds Ga-68 (t1/2: 67.7 min) at much lower temperature, thus allowing for quick access to "ready-for-injection" [68Ga]Ga-tracers in hospitals. We herein introduce [68Ga]Ga-DATA5m-LM4 for PET/CT imaging of SST2R-positive human tumors. LM4 was obtained by 4Pal3/Tyr3-substitution in the known SST2R antagonist LM3 (H-DPhe-c[DCys-Tyr-DAph(Cbm)-Lys-Thr-Cys]-DTyr-NH2) and DATA5m was coupled at the N-terminus for labeling with radiogallium (Ga-67/68). [67Ga]Ga-DATA5m-LM4 was evaluated in HEK293-SST2R cells and mice models in a head-to-head comparison with [67Ga]Ga-DOTA-LM3. Clinical grade [68Ga]Ga-DATA5m-LM4 was prepared and injected in a neuroendocrine tumor (NET) patient for PET/CT imaging. DATA5m-LM4 displayed high SST2R binding affinity. [67Ga]Ga-DATA5m-LM4 showed markedly higher uptake in HEK293-SST2R cells versus [67Ga]Ga-DOTA-LM3 and was stable in vivo. In HEK293-SST2R xenograft-bearing mice, it achieved longer tumor retention and less kidney uptake than [67Ga]Ga-DOTA-LM3. [68Ga]Ga-DATA5m-LM4 accurately visualized tumor lesions with high contrast on PET/CT. In short, [68Ga]Ga-DATA5m-LM4 has shown excellent prospects for the PET/CT diagnosis of SST2R-positive tumors, further highlighting the benefits of Ga-68 labeling in a hospital environment via the DATA5m-chelator route.
Collapse
Affiliation(s)
| | - Berthold A. Nock
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, GR-15310 Athens, Greece
| | - Lukas Greifenstein
- CURANOSTICUM Wiesbaden-Frankfurt, DKD Helios Klinik, D-65191 Wiesbaden, Germany
| | - Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, DKD Helios Klinik, D-65191 Wiesbaden, Germany
| | - Frank Roesch
- Department Chemie, Standort TRIGA, Johannes Gutenberg-Universität Mainz, D-55126 Mainz, Germany
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, GR-15310 Athens, Greece
- Correspondence: ; Tel.: +30-210-650-3908 (ext. 3891)
| |
Collapse
|