1
|
Fonseca AI, Sereno J, Almeida S, Ferreira H, Hrynchak I, Falcão A, Alves F, Gomes C, Abrunhosa AJ. Unveiling the potential of copper-61 vs. gallium-68 for SSTR PET imaging. Eur J Nucl Med Mol Imaging 2025; 52:2671-2684. [PMID: 39909885 PMCID: PMC12119765 DOI: 10.1007/s00259-025-07116-2] [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: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE In recent years, copper-61 has attracted considerable attention from both physicists and radiochemists due to its favorable physical decay properties for PET imaging and its ease of production at any cyclotron center producing [18F]FDG. The aim of this study was to evaluate the potential of 61Cu-based radiopharmaceuticals for PET imaging of NETs, as an alternative to the commonly used gallium-68. METHODS Copper-61 was produced by irradiation of natural zinc liquid targets, followed by post-processing. In vitro evaluation of 61Cu- and 68Ga-labeled SST analogues was performed in SSTR positive AR42J tumor cells. PET/MRI was carried out in mice bearing AR42J subcutaneous tumors. RESULTS High molar activity [61Cu]Cu-DOTA-TATE and [61Cu]Cu-NOTA-TATE were successfully prepared with a radiochemical purity of over 95% and were shown to be stable for at least 6 h after the EOS. Both 61Cu- and 68Ga-labeled SST analogues exhibited high cellular uptake, with residual uptake when blocked with an excessive amount of peptide precursor. [61Cu]Cu-NOTA-TATE showed the highest tumor uptake at 1 h p.i. (13.25 ± 1.86%ID/g) and the tumor-to-non-tumor ratio increased from 1 h to 4 h p.i. At the later time point, tumor visualization improved compared to 1 h p.i. Moreover, preclinical PET/MR images demonstrated that [61Cu]Cu-NOTA-TATE has a more favorable biodistribution and imaging properties than [61Cu]Cu-DOTA-TATE, with the extended PET imaging window providing a clear advantage of [61Cu]Cu-NOTA-TATE over its gallium-68 analogues. CONCLUSION [61Cu]Cu-NOTA-TATE showed similar biodistribution and pharmacokinetics to [68Ga]Ga-DOTA-TATE at 1 h p.i., while demonstrating superior imaging characteristics for late PET imaging. These findings demonstrate that [61Cu]Cu-NOTA-TATE holds promising characteristics for improving the detection of NETs with increased translational potential.
Collapse
Affiliation(s)
- A I Fonseca
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
| | - J Sereno
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
| | - S Almeida
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
| | - H Ferreira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, Portugal
| | - I Hrynchak
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
| | - A Falcão
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - F Alves
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
- ESTeSC - Coimbra Health School, Instituto Politécnico Coimbra, Coimbra, Portugal
| | - C Gomes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, Portugal
| | - A J Abrunhosa
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
2
|
Rudd SE, Van Zuylekom J, Cullinane C, Blyth BJ, Donnelly PS. Potential theranostics of breast cancer with copper-64/67 sarcophagine-trastuzumab. Chem Sci 2025; 16:3998-4005. [PMID: 39898305 PMCID: PMC11783091 DOI: 10.1039/d4sc06969b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
Over-expression of Human Epidermal Growth Factor Receptor 2 (HER2) is associated with a significant proportion of breast cancers. Targeting HER2 is possible with a monoclonal antibody called trastuzumab but metastatic HER2 positive tumours can develop resistance to this treatment. One approach to develop more potent therapeutic agents which retain the selectivity of trastuzumab is to attach a β- emitting radionuclide, such as copper-67, to the antibody for radioimmunotherapy. It is also possible to attach β+ emitting copper-64 to antibodies for diagnostic imaging with positron emission tomography (PET). In this work, a cage amine sarcophagine (Sar) chelator is conjugated to trastuzumab to give Sar-trastuzumab which can be radiolabelled with either copper-64 or copper-67 at room temperature in minutes to give [64Cu]CuSar-trastuzumab and [67Cu]CuSar-trastuzumab respectively. The diagnostic imaging potential of [64Cu]CuSar-trastuzumab was evaluated in mice bearing HER2+ SKOV-3 tumours showing that the tracer has very high tumour uptake and retention 48 hours after injection. The copper-67 variant, [67Cu]CuSar-trastuzumab, was highly therapeutically efficacious in the same tumour model with no signs of radiotoxicity. The combination of diagnostic PET imaging with [64Cu]CuSar-trastuzumab to guide radionuclide therapy with [67Cu]CuSar-trastzumab has significant potential for theranostic treatment of breast cancer and other HER2+ disease that has become resistant to conventional immunotherapy.
Collapse
Affiliation(s)
- Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Melbourne Victoria 3010 Australia
| | - Jessica Van Zuylekom
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre 305 Grattan St Melbourne Victoria 3000 Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne Parkville Victoria 3010 Australia
| | - Carleen Cullinane
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre 305 Grattan St Melbourne Victoria 3000 Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne Parkville Victoria 3010 Australia
| | - Benjamin J Blyth
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre 305 Grattan St Melbourne Victoria 3000 Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne Parkville Victoria 3010 Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Melbourne Victoria 3010 Australia
| |
Collapse
|
3
|
Kręcisz P, Stefańska K, Studziński J, Pitucha M, Czylkowska A, Szymański P. Radiocopper in Radiopharmacy and Medical Use: Current Status and Perspective. J Med Chem 2025; 68:2356-2376. [PMID: 39895089 PMCID: PMC11831595 DOI: 10.1021/acs.jmedchem.4c02885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Of the 32 known copper isotopes, some have interesting properties for nuclear medicine, for example the short-lived 60Cu, 61Cu, 62Cu, the moderate long-lived 64Cu and the long-lived 67Cu. Due to their emission properties, copper isotopes are suitable for both imaging diagnostics (60Cu, 61Cu, 62Cu, 64Cu) and targeted radiotherapy (64Cu and 67Cu). As their chemical properties are virtually identical, a single radiopharmaceutical structure can be labeled with different isotopes, depending on the clinical application. This, combined with the ability to combine radioisotopes with different nuclear properties with the same ligand, makes them extremely versatile. The purpose of this review is to introduce the world of copper radiopharmaceuticals and to summarize recent advances in methods for producing copper radioisotopes and the preclinical research of radiopharmaceuticals labeled with copper radioisotopes.
Collapse
Affiliation(s)
- Paweł Kręcisz
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Katarzyna Stefańska
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Jakub Studziński
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Monika Pitucha
- Independent
Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Agnieszka Czylkowska
- Institute
of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Paweł Szymański
- Department
of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty
of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
- Department
of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| |
Collapse
|
4
|
Nazar AK, Basu S. Radiolabeled Somatostatin Analogs for Cancer Imaging. Semin Nucl Med 2024; 54:914-940. [PMID: 39122608 DOI: 10.1053/j.semnuclmed.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024]
Abstract
Somatostatin receptors (SSTR) are expressed by many tumours especially those related to neuro-endocrine origin and molecular functional imaging of SSTR expression using radiolabelled somatostatin analogs have revolutionized imaging of patients with these group of malignancies. Coming a long way from the first radiolabelled somatostatin analog 123I-Tyr-3-octreotide, there has been significant developments in terms of radionuclides used, the ligands and somatostatin derivatives. 111In-Pentetreotide extensively employed for imaging NETs at the beginning has now been replaced by 68Ga-SSA based PET-CT. SSA-PET/CT performs superior to conventional imaging modalities and has evolved in the mainframe for NET imaging. The advantages were multiple: (i) superior spatial resolution of PET versus SPECT, (ii) quantitative capabilities of PET aiding in disease activity and treatment response monitoring with better precision, (iii) shorter scan time and (iv) less patient exposure to radiation. The modality is indicated for staging, detecting the primary in CUP-NETs, restaging, treatment planning (along with FDG: the concept of dual-tracer PET-CT) as well as treatment response evaluation and follow-up of NETs. SSA PET/CT has also been incorporated in the guidelines for imaging of Pheochromocytoma-Paraganglioma, Medullary carcinoma thyroid, Meningioma and Tumor induced osteomalacia. At present, there is rising interest on (a) 18F-labelled SSA, (b) 64Cu-labelled SSA, and (c) somatostatin antagonists. 18F offers excellent imaging properties, 64Cu makes delayed imaging feasible which has implications in dosimetry and SSTR antagonists bind with the SST receptors with high affinity and specificity, providing high contrast images with less background, which can be translated to theranostics effectively. SSTR have been demonstrated in non-neuroendocrine tumours as well in the peer-reviewed literature, with studies demonstrating the potential of SSA PET/CT in Neuroblastoma, Nasopharyngeal carcinoma, carcinoma prostate (neuroendocrine differentiation) and lymphoma. This review will focus on the currently available SSAs and their history, different SPECT/PET agents, SSTR antagonists, comparison between the various imaging tracers, and their utility in both neuroendocrine and non-neuroendocrine tumors.
Collapse
Affiliation(s)
- Aamir K Nazar
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai.
| |
Collapse
|
5
|
Mallak N, Yilmaz B, Meyer C, Winters C, Mench A, Jha AK, Prasad V, Mittra E. Theranostics in Neuroendocrine Tumors: Updates and Emerging Technologies. Curr Probl Cancer 2024; 52:101129. [PMID: 39232443 DOI: 10.1016/j.currproblcancer.2024.101129] [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: 12/17/2023] [Accepted: 05/22/2024] [Indexed: 09/06/2024]
Abstract
Advancements in somatostatin receptor (SSTR) targeted imaging and treatment of well-differentiated neuroendocrine tumors (NETs) have revolutionized the management of these tumors. This comprehensive review delves into the current practice, discussing the use of the various FDA-approved SSTR-agonist PET tracers and the predictive imaging biomarkers, and elaborating on Lu177-DOTATATE peptide receptor radionuclide therapy (PRRT) including the evolving areas of post-therapy imaging practices, PRRT retreatment, and the potential role of dosimetry in optimizing patient treatments. The future directions sections highlight ongoing research on investigational PET imaging radiotracers, future prospects in alpha particle therapy, and combination therapy strategies.
Collapse
Affiliation(s)
- Nadine Mallak
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Burcak Yilmaz
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Catherine Meyer
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Celeste Winters
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Anna Mench
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Abhinav K Jha
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA; Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Vikas Prasad
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Erik Mittra
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA.
| |
Collapse
|
6
|
Niego B, Jupp B, Zia NA, Xu R, Jap E, Ezeani M, Noor A, Donnelly PS, Hagemeyer CE, Alt K. Molecular Imaging of Diffuse Cardiac Fibrosis with a Radiotracer That Targets Proteolyzed Collagen IV. Radiol Cardiothorac Imaging 2024; 6:e230098. [PMID: 38512024 PMCID: PMC11056764 DOI: 10.1148/ryct.230098] [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: 04/05/2023] [Revised: 11/19/2023] [Accepted: 01/22/2024] [Indexed: 03/22/2024]
Abstract
Purpose To develop an approach for in vivo detection of interstitial cardiac fibrosis using PET with a peptide tracer targeting proteolyzed collagen IV (T-peptide). Materials and Methods T-peptide was conjugated to the copper chelator MeCOSar (chemical name, 5-(8-methyl-3,6,10,13,16,19-hexaaza-bicyclo[6.6.6]icosan-1-ylamino)-5-oxopentanoic acid) and radiolabeled with copper 64 (64Cu). PET/CT scans were acquired following intravenous delivery of 64Cu-T-peptide-MeCOSar (0.25 mg/kg; 18 MBq ± 2.7 [SD]) to male transgenic mice overexpressing β2-adrenergic receptors with intermediate (7 months of age; n = 4 per group) to severe (10 months of age; n = 11 per group) cardiac fibrosis and their wild-type controls. PET scans were also performed following coadministration of the radiolabeled probe with nonlabeled T-peptide in excess to confirm binding specificity. PET data were analyzed by t tests for static scans and analysis of variance tests (one- or two-way) for dynamic scans. Results PET/CT scans revealed significantly elevated (2.24-4.26-fold; P < .05) 64Cu-T-peptide-MeCOSar binding in the fibrotic hearts of aged transgenic β2-adrenergic receptor mice across the entire 45-minute acquisition period compared with healthy controls. The cardiac tracer accumulation and presence of diffuse cardiac fibrosis in older animals were confirmed by gamma counting (P < .05) and histologic evaluation, respectively. Coadministration of a nonradiolabeled probe in excess abolished the elevated radiotracer binding in the aged transgenic hearts. Importantly, PET tracer accumulation was also detected in younger (7 months of age) transgenic mice with intermediate cardiac fibrosis, although this was only apparent from 20 minutes following injection (1.6-2.2-fold binding increase; P < .05). Conclusion The T-peptide PET tracer targeting proteolyzed collagen IV provided a sensitive and specific approach of detecting diffuse cardiac fibrosis at varying degrees of severity in a transgenic mouse model. Keywords: Diffuse Cardiac Fibrosis, Molecular Peptide Probe, Molecular Imaging, PET/CT © RSNA, 2024.
Collapse
Affiliation(s)
| | | | - Nicholas A. Zia
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | - Rong Xu
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | - Edwina Jap
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | - Martin Ezeani
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | - Asif Noor
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | - Paul S. Donnelly
- From the NanoBiotechnology Laboratory (B.N., R.X., M.E., C.E.H.) and
NanoTheranostics Laboratory (E.J., K.A.), Australian Centre for Blood Diseases,
Central Clinical School, Monash University, Melbourne, VIC 3004, Australia;
Department of Neuroscience, Central Clinical School, Monash University,
Melbourne, Australia (B.J.); and School of Chemistry and Bio21 Molecular Science
and Biotechnology Institute, University of Melbourne, Melbourne, Australia
(N.A.Z., A.N., P.S.D.)
| | | | | |
Collapse
|
7
|
Lin W, Fonseca Cabrera GO, Aluicio-Sarduy E, Barnhart TE, Mixdorf JC, Li Z, Wu Z, Engle JW. Radiolabeling Diaminosarcophagine with Cyclotron-Produced Cobalt-55 and [ 55Co]Co-NT-Sarcage as a Proof of Concept in a Murine Xenograft Model. Bioconjug Chem 2024; 35:412-418. [PMID: 38411531 PMCID: PMC10954389 DOI: 10.1021/acs.bioconjchem.4c00043] [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] [Indexed: 02/28/2024]
Abstract
Cobalt-sarcophagine complexes exhibit high kinetic inertness under various stringent conditions, but there is limited literature on radiolabeling and in vivo positron emission tomography (PET) imaging using no carrier added 55Co. To fill this gap, this study first investigates the radiolabeling of DiAmSar (DSar) with 55Co, followed by stability evaluation in human serum and EDTA, pharmacokinetics in mice, and a direct comparison with [55Co]CoCl2 to assess differences in pharmacokinetics. Furthermore, the radiolabeling process was successfully used to generate the NTSR1-targeted PET agent [55Co]Co-NT-Sarcage (a DSar-functionalized SR142948 derivative) and administered to HT29 tumor xenografted mice. The [55Co]Co-DSar complex can be formed at 37 °C with purity and stability suitable for preclinical in vivo radiopharmaceutical applications, and [55Co]Co-NT-Sarcage demonstrated prominent tumor uptake with a low background signal. In a direct comparison with [64Cu]Cu-NT-Sarcage, [55Co]Co-NT-Sarcage achieved a higher tumor-to-liver ratio but with overall similar biodistribution profile. These results demonstrate that Sar would be a promising chelator for constructing Co-based radiopharmaceuticals including 55Co for PET and 58mCo for therapeutic applications.
Collapse
Affiliation(s)
- Wilson Lin
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, United States
| | - German Oscar Fonseca Cabrera
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, United States
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, United States
| | - Jason C. Mixdorf
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, United States
| | - Zibo Li
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Zhanhong Wu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, United States
- Department of Radiology, University of Wisconsin, 600 Highland Ave., Madison, WI, 53792, United States
| |
Collapse
|
8
|
Rowan JA, Rudd SE, Ganio K, McDevitt CA, White JM, Donnelly PS. Copper(II) Complexes of 2,2'- Bisdipyrrins: Synthesis, Characterization, Cell Uptake, and Radiolabeling with Copper-64. Inorg Chem 2023; 62:20666-20676. [PMID: 37552883 DOI: 10.1021/acs.inorgchem.3c01551] [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: 08/10/2023]
Abstract
Complexes prepared with positron-emitting copper-64 are of interest as imaging agents for positron emission tomography (PET). This work investigates the potential of using acyclic tetrapyrrolic 2,2'-bisdipyrrins as ligands to prepare charge-neutral, lipophilic, cell-permeable, redox active complexes with positron-emitting copper-64. The synthesis and characterization of a series of tetrapyrrolic 2,2'-bisdipyrrin copper(II) complexes are reported. Four 2,2'-bisdipyrrin copper(II) complexes were prepared with different functional groups in the meso-position of the ligands. Two of the new copper(II) complexes, one palladium(II) complex, and one nickel(II) complex were characterized by X-ray crystallography, which demonstrated that the copper(II) is in a distorted square planar environment. An investigation of the electrochemical properties of the complexes by cyclic voltammetry revealed that the complexes undergo multiple quasi-reversible processes. A comparison of the cyclic voltammetry of the copper complexes with their palladium(II) analogues suggests that these redox processes are ligand-based and not metal-based. The copper(II) complexes are cell-permeable in A431 mammalian cells and are nontoxic at concentrations of 50 μM. The ligands can be radiolabeled with copper-64 at room temperature.
Collapse
Affiliation(s)
- Jacob A Rowan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Katherine Ganio
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Christopher A McDevitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Jonathan M White
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
9
|
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: 37] [Impact Index Per Article: 18.5] [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
|
10
|
Echavidre W, Fagret D, Faraggi M, Picco V, Montemagno C. Recent Pre-Clinical Advancements in Nuclear Medicine: Pioneering the Path to a Limitless Future. Cancers (Basel) 2023; 15:4839. [PMID: 37835533 PMCID: PMC10572076 DOI: 10.3390/cancers15194839] [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: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The theranostic approach in oncology holds significant importance in personalized medicine and stands as an exciting field of molecular medicine. Significant achievements have been made in this field in recent decades, particularly in treating neuroendocrine tumors using 177-Lu-radiolabeled somatostatin analogs and, more recently, in addressing prostate cancer through prostate-specific-membrane-antigen targeted radionuclide therapy. The promising clinical results obtained in these indications paved the way for the further development of this approach. With the continuous discovery of new molecular players in tumorigenesis, the development of novel radiopharmaceuticals, and the potential combination of theranostics agents with immunotherapy, nuclear medicine is poised for significant advancements. The strategy of theranostics in oncology can be categorized into (1) repurposing nuclear medicine agents for other indications, (2) improving existing radiopharmaceuticals, and (3) developing new theranostics agents for tumor-specific antigens. In this review, we provide an overview of theranostic development and shed light on its potential integration into combined treatment strategies.
Collapse
Affiliation(s)
- William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, CHU Grenoble Alpes, Inserm, 38000 Grenoble, France;
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| |
Collapse
|
11
|
Haq A, Rayamajhi S, Ponisio MR, Prasad V. New horizon of radiopharmaceuticals in management of neuroendocrine tumors. Best Pract Res Clin Endocrinol Metab 2023; 37:101797. [PMID: 37468403 DOI: 10.1016/j.beem.2023.101797] [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] [Indexed: 07/21/2023]
Abstract
Neuroendocrine neoplasms are rare and heterogenous group of tumors with varying degrees of clinical presentations and involvement of multiple organ systems in the body. In the modern clinical practice somatostatin receptor molecular imaging and targeted radioligand therapy plays a vital role in the diagnosis and management of the disease. Several new and promising radiotracers for NET imaging and theranostics, belonging to various groups and classes are being studied and investigated. This exponential growth of radiotracers poses concerns about the indication, clinical benefit, and safety profile of the agents. We discuss the basis behind these radiotracers clinical use, receptor targeting and intra and inter tumor heterogeneity. Furthermore, role of dual tracer imaging, combination therapy and potential applications of dosimetry in predicting treatment outcome and safety profile is reviewed. Individualized precision medicine with better tumor characterization, maximum therapeutic benefit and minimum toxicity is the way forward for future medicine.
Collapse
Affiliation(s)
- Adeel Haq
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, MO, United States.
| | - Sampanna Rayamajhi
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, MO, United States
| | - Maria Rosana Ponisio
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, MO, United States
| | - Vikas Prasad
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
12
|
Gervasoni S, Öztürk I, Guccione C, Bosin A, Ruggerone P, Malloci G. Interaction of Radiopharmaceuticals with Somatostatin Receptor 2 Revealed by Molecular Dynamics Simulations. J Chem Inf Model 2023; 63:4924-4933. [PMID: 37466559 PMCID: PMC10428218 DOI: 10.1021/acs.jcim.3c00712] [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: 05/12/2023] [Indexed: 07/20/2023]
Abstract
The development of drugs targeting somatostatin receptor 2 (SSTR2), generally overexpressed in neuroendocrine tumors, is focus of intense research. A few molecules in conjugation with radionuclides are in clinical use for both diagnostic and therapeutic purposes. These radiopharmaceuticals are composed of a somatostatin analogue biovector conjugated to a chelator moiety bearing the radionuclide. To date, despite valuable efforts, a detailed molecular-level description of the interaction of radiopharmaceuticals in complex with SSTR2 has not yet been accomplished. Therefore, in this work, we carefully analyzed the key dynamical features and detailed molecular interactions of SSTR2 in complex with six radiopharmaceutical compounds selected among the few already in use (64Cu/68Ga-DOTATATE, 68Ga-DOTATOC, 64Cu-SARTATE) and some in clinical development (68Ga-DOTANOC, 64Cu-TETATATE). Through molecular dynamics simulations and exploiting recently available structures of SSTR2, we explored the influence of the different portions of the compounds (peptide, radionuclide, and chelator) in the interaction with the receptor. We identified the most stable binding modes and found distinct interaction patterns characterizing the six compounds. We thus unveiled detailed molecular interactions crucial for the recognition of this class of radiopharmaceuticals. The microscopically well-founded analysis presented in this study provides guidelines for the design of new potent ligands targeting SSTR2.
Collapse
Affiliation(s)
| | | | - Camilla Guccione
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Andrea Bosin
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| |
Collapse
|
13
|
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
|
14
|
Brown AM, Butman JL, Lengacher R, Vargo NP, Martin KE, Koller A, Śmiłowicz D, Boros E, Robinson JR. N, N-Alkylation Clarifies the Role of N- and O-Protonated Intermediates in Cyclen-Based 64Cu Radiopharmaceuticals. Inorg Chem 2023; 62:1362-1376. [PMID: 36490364 DOI: 10.1021/acs.inorgchem.2c02907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Radioisotopes of Cu, such as 64Cu and 67Cu, are alluring targets for imaging (e.g., positron emission tomography, PET) and radiotherapeutic applications. Cyclen-based macrocyclic polyaminocarboxylates are one of the most frequently examined bifunctional chelators in vitro and in vivo, including the FDA-approved 64Cu radiopharmaceutical, Cu(DOTATATE) (Detectnet); however, connections between the structure of plausible reactive intermediates and their stability under physiologically relevant conditions remain to be established. In this study, we share the synthesis of a cyclen-based, N,N-alkylated spirocyclic chelate, H2DO3AC4H8, which serves as a model for N-protonation. Our combined experimental (in vitro and in vivo) and computational studies unravel complex pH-dependent speciation and enable side-by-side comparison of N- and O-protonated species of relevant 64Cu radiopharmaceuticals. Our studies suggest that N-protonated species are not inherently unstable species under physiological conditions and demonstrate the potential of N,N-alkylation as a tool for the rational design of future radiopharmaceuticals.
Collapse
Affiliation(s)
- Alexander M Brown
- Department of Chemistry, Brown University, Providence, Rhode Island02912, United States
| | - Jana L Butman
- Department of Chemistry, Brown University, Providence, Rhode Island02912, United States
| | - Raphael Lengacher
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York11794, United States
| | - Natasha P Vargo
- Department of Chemistry, Brown University, Providence, Rhode Island02912, United States
| | - Kirsten E Martin
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York11794, United States
| | - Angus Koller
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York11794, United States
| | - Dariusz Śmiłowicz
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York11794, United States
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York11794, United States
| | - Jerome R Robinson
- Department of Chemistry, Brown University, Providence, Rhode Island02912, United States
| |
Collapse
|
15
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
16
|
Calatayud DG, Neophytou S, Nicodemou E, Giuffrida SG, Ge H, Pascu SI. Nano-Theranostics for the Sensing, Imaging and Therapy of Prostate Cancers. Front Chem 2022; 10:830133. [PMID: 35494646 PMCID: PMC9039169 DOI: 10.3389/fchem.2022.830133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/16/2022] [Indexed: 01/28/2023] Open
Abstract
We highlight hereby recent developments in the emerging field of theranostics, which encompasses the combination of therapeutics and diagnostics in a single entity aimed for an early-stage diagnosis, image-guided therapy as well as evaluation of therapeutic outcomes of relevance to prostate cancer (PCa). Prostate cancer is one of the most common malignancies in men and a frequent cause of male cancer death. As such, this overview is concerned with recent developments in imaging and sensing of relevance to prostate cancer diagnosis and therapeutic monitoring. A major advantage for the effective treatment of PCa is an early diagnosis that would provide information for an appropriate treatment. Several imaging techniques are being developed to diagnose and monitor different stages of cancer in general, and patient stratification is particularly relevant for PCa. Hybrid imaging techniques applicable for diagnosis combine complementary structural and morphological information to enhance resolution and sensitivity of imaging. The focus of this review is to sum up some of the most recent advances in the nanotechnological approaches to the sensing and treatment of prostate cancer (PCa). Targeted imaging using nanoparticles, radiotracers and biomarkers could result to a more specialised and personalised diagnosis and treatment of PCa. A myriad of reports has been published literature proposing methods to detect and treat PCa using nanoparticles but the number of techniques approved for clinical use is relatively small. Another facet of this report is on reviewing aspects of the role of functional nanoparticles in multimodality imaging therapy considering recent developments in simultaneous PET-MRI (Positron Emission Tomography-Magnetic Resonance Imaging) coupled with optical imaging in vitro and in vivo, whilst highlighting feasible case studies that hold promise for the next generation of dual modality medical imaging of PCa. It is envisaged that progress in the field of imaging and sensing domains, taken together, could benefit from the biomedical implementation of new synthetic platforms such as metal complexes and functional materials supported on organic molecular species, which can be conjugated to targeting biomolecules and encompass adaptable and versatile molecular architectures. Furthermore, we include hereby an overview of aspects of biosensing methods aimed to tackle PCa: prostate biomarkers such as Prostate Specific Antigen (PSA) have been incorporated into synthetic platforms and explored in the context of sensing and imaging applications in preclinical investigations for the early detection of PCa. Finally, some of the societal concerns around nanotechnology being used for the detection of PCa are considered and addressed together with the concerns about the toxicity of nanoparticles–these were aspects of recent lively debates that currently hamper the clinical advancements of nano-theranostics. The publications survey conducted for this review includes, to the best of our knowledge, some of the most recent relevant literature examples from the state-of-the-art. Highlighting these advances would be of interest to the biomedical research community aiming to advance the application of theranostics particularly in PCa diagnosis and treatment, but also to those interested in the development of new probes and methodologies for the simultaneous imaging and therapy monitoring employed for PCa targeting.
Collapse
Affiliation(s)
- David G. Calatayud
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Department of Electroceramics, Instituto de Ceramica y Vidrio - CSIC, Madrid, Spain
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| | - Sotia Neophytou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Eleni Nicodemou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | | | - Haobo Ge
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Sofia I. Pascu
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Centre of Therapeutic Innovations, University of Bath, Bath, United Kingdom
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| |
Collapse
|
17
|
Abstract
Artificial receptors able to recognise biologically relevant molecules or ions have gained interest in the chemical community because they offer a plethora of posibilities. Molecular cage compounds are polycyclic compounds with a cavity designed for the encapsulation of guest species. Once inside the host cavity, the substrate can be transported through membranes and protected from the action of enzymes or other reactive species, thus offering the possibility of interfering with biological systems. Commonly, enzymes have been an inspiration for chemists in the search and design of defined cavities for different purposes. However, the chemical preparation of molecular cages has struggled with many synthetic challenges but this effort is worthwhile as they are a very promising tool for many applications ranging from sensing, delivery, purification or even promotion of/prevention from chemical modifications. Since the early reports at the end of the 60s, this field has experienced a growing interest; this review summarises the progress in the preparation and study of cage-like compounds highlighting their importance in biological applications.
Collapse
Affiliation(s)
- Lucía Tapia
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Ignacio Alfonso
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain.
| | - Jordi Solà
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia, IQAC-CSIC, Jordi Girona 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
18
|
Choiński J, Łyczko M. Prospects for the production of radioisotopes and radiobioconjugates for theranostics. BIO-ALGORITHMS AND MED-SYSTEMS 2021. [DOI: 10.1515/bams-2021-0136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract
The development of diagnostic methods in medicine as well as the progress in the synthesis of biologically active compounds allows the use of selected radioisotopes for the simultaneous diagnosis and treatment of diseases, especially cancerous ones, in patients. This approach is called theranostic. This review article includes chemical and physical characterization of chosen theranostic radioisotopes and their compounds that are or could be useful in nuclear medicine.
Collapse
Affiliation(s)
| | - Monika Łyczko
- Institute of Nuclear Chemistry and Technology , Warsaw , Poland
| |
Collapse
|
19
|
Farleigh M, Pham TT, Yu Z, Kim J, Sunassee K, Firth G, Forte N, Chudasama V, Baker JR, Long NJ, Rivas C, Ma MT. New Bifunctional Chelators Incorporating Dibromomaleimide Groups for Radiolabeling of Antibodies with Positron Emission Tomography Imaging Radioisotopes. Bioconjug Chem 2021; 32:1214-1222. [PMID: 33724798 PMCID: PMC8299457 DOI: 10.1021/acs.bioconjchem.0c00710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Indexed: 01/23/2023]
Abstract
Positron Emission Tomography (PET) imaging with antibody-based contrast agents frequently uses the radioisotopes [64Cu]Cu2+ and [89Zr]Zr4+. The macrobicyclic chelator commonly known as sarcophagine (sar) is ideal for labeling receptor-targeted biomolecules with [64Cu]Cu2+. The siderophore chelator, desferrioxamine-B (dfo), has been widely used to incorporate [89Zr]Zr4+ into antibodies. Here, we describe new bifunctional chelators of sar and dfo: these chelators have been functionalized with dibromomaleimides (dbm), that enable site-specific and highly stable attachment of molecular cargoes to reduced, solvent-accessible, interstrand native disulfide groups. The new sar-dbm and dfo-dbm derivatives can be easily conjugated with the IgG antibody trastuzumab via reaction with reduced interstrand disulfide groups to give site-specifically modified dithiomaleamic acid (dtm) conjugates, sar-dtm-trastuzumab and dfo-dtm-trastuzumab, in which interstrand disulfides are rebridged covalently with a small molecule linker. Both sar- and dfo-dtm-trastuzumab conjugates have been radiolabeled with [64Cu]Cu2+ and [89Zr]Zr4+, respectively, in near quantitative radiochemical yield (>99%). Serum stability studies, in vivo PET imaging, and biodistribution analyses using these radiolabeled immunoconjugates demonstrate that both [64Cu]Cu-sar-dtm-trastuzumab and [89Zr]Zr-dfo-dtm-trastuzumab possess high stability in biological milieu. Dibromomaleimide technology can be easily applied to enable stable, site-specific attachment of radiolabeled chelators, such as sar and dfo, to native interstrand disulfide regions of antibodies, enabling tracking of antibodies with PET imaging.
Collapse
Affiliation(s)
- Matthew Farleigh
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Truc Thuy Pham
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Zilin Yu
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Jana Kim
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Kavitha Sunassee
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - George Firth
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Nafsika Forte
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - James R. Baker
- Department
of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, U.K.
| | - Nicholas J. Long
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, London W12 0BZ, U.K.
| | - Charlotte Rivas
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Michelle T. Ma
- School
of Biomedical Engineering and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| |
Collapse
|
20
|
Rudd SE, Van Zuylekom JK, Raicevic A, Pearce LA, Cullinane C, Williams CC, Adams TE, Hicks RJ, Donnelly PS. Enzyme mediated incorporation of zirconium-89 or copper-64 into a fragment antibody for same day imaging of epidermal growth factor receptor. Chem Sci 2021; 12:9004-9016. [PMID: 34276928 PMCID: PMC8261882 DOI: 10.1039/d1sc01422f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022] Open
Abstract
Identification of tumors which over-express Epidermal Growth Factor Receptor (EGFR) is important in selecting patients for anti-EGFR therapies. Enzymatic bioconjugation was used to introduce positron-emitting radionuclides (89Zr, 64Cu) into an anti-EGFR antibody fragment for Positron Emission Tomography (PET) imaging the same day as injection. A monovalent antibody fragment with high affinity for EGFR was engineered to include a sequence that is recognized by the transpeptidase sortase A. Two different metal chelators, one for 89ZrIV and one for 64CuII, were modified with a N-terminal glycine to enable them to act as substrates in sortase A mediated bioconjugation to the antibody fragment. Both fragments provided high-quality PET images of EGFR positive tumors in a mouse model at 3 hours post-injection, a significant advantage when compared to radiolabeled full antibodies that require several days between injection of the tracer and imaging. The use of enzymatic bioconjugation gives reproducible homogeneous products with the metal complexes selectively installed on the C-terminus of the antibody potentially simplifying regulatory approval.
Collapse
Affiliation(s)
- Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science, Biotechnology Institute University of Melbourne Parkville Victoria 3010 Australia
| | | | - Anna Raicevic
- CSIRO Manufacturing Parkville Victoria 3052 Australia
| | | | - Carleen Cullinane
- Research Division, Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne Parkville Victoria Australia
| | | | | | - Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne Parkville Victoria Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science, Biotechnology Institute University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
21
|
Noor A, Van Zuylekom JK, Rudd SE, Roselt PD, Haskali MB, Yan E, Wheatcroft M, Hicks RJ, Cullinane C, Donnelly PS. Imaging Somatostatin Positive Tumors with Tyr 3-Octreotate/Octreotide Conjugated to Desferrioxamine B Squaramide Radiolabeled with either Zirconium-89 or Gallium-68. Bioconjug Chem 2021; 32:1192-1203. [PMID: 33788556 DOI: 10.1021/acs.bioconjchem.1c00109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Radiolabeled derivatives of Tyr3-octreotide and Tyr3-octreotate, synthetic analogues of the peptide hormone somatostatin, can be used for positron emission tomography (PET) imaging of somatostatin receptor expression in neuroendocrine tumors. In this work, a squaramide ester derivative of desferrioxamine B (H3DFOSq) was used attach either Tyr3-octreotide or Tyr3-octreotate to the metal binding ligand to give H3DFOSq-TIDE and H3DFOSq-TATE. These new peptide-H3DFOSq conjugates form stable complexes with either of the positron-emitting radionuclides gallium-68 (t1/2 = 68 min) or zirconium-89 (t1/2 = 3.3 days). The new complexes were evaluated in an AR42J xenograft model that has endogenous expression of SSTR2. All four agents displayed good tumor uptake and produced high-quality PET images. For both radionuclides, the complexes formed with H3DFOSq-TATE performed better, with higher tumor uptake and retention than the complexes formed with H3DFOSq-TIDE. The versatile ligands presented here can be radiolabeled with either gallium-68 or zirconium-89 at room temperature. The long radioactive half-life of zirconium-89 makes distribution of pre-synthesized tracers produced to certified standards feasible and could increase the number of clinical centers that can perform diagnostic PET imaging of neuroendocrine tumors.
Collapse
Affiliation(s)
- Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter D Roselt
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Mohammad B Haskali
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Eddie Yan
- Telix Pharmaceuticals Limited, Suite 401, 55 Flemington Road, North Melbourne, Victoria 3051, Australia
| | - Michael Wheatcroft
- Telix Pharmaceuticals Limited, Suite 401, 55 Flemington Road, North Melbourne, Victoria 3051, Australia
| | - Rodney J Hicks
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Carleen Cullinane
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
22
|
Dearling JLJ, van Dam EM, Harris MJ, Packard AB. Detection and therapy of neuroblastoma minimal residual disease using [ 64/67Cu]Cu-SARTATE in a preclinical model of hepatic metastases. EJNMMI Res 2021; 11:20. [PMID: 33630166 PMCID: PMC7907331 DOI: 10.1186/s13550-021-00763-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/15/2021] [Indexed: 12/27/2022] Open
Abstract
Background A major challenge to the long-term success of neuroblastoma therapy is widespread metastases that survive initial therapy as minimal residual disease (MRD). The SSTR2 receptor is expressed by most neuroblastoma tumors making it an attractive target for molecularly targeted radionuclide therapy. SARTATE consists of octreotate, which targets the SSTR2 receptor, conjugated to MeCOSar, a bifunctional chelator with high affinity for copper. Cu-SARTATE offers the potential to both detect and treat neuroblastoma MRD by using [64Cu]Cu-SARTATE to detect and monitor the disease and [67Cu]Cu-SARTATE as the companion therapeutic agent. In the present study, we tested this theranostic pair in a preclinical model of neuroblastoma MRD. An intrahepatic model of metastatic neuroblastoma was established using IMR32 cells in nude mice. The biodistribution of [64Cu]Cu-SARTATE was measured using small-animal PET and ex vivo tissue analysis. Survival studies were carried out using the same model: mice (6–8 mice/group) were given single doses of saline, or 9.25 MBq (250 µCi), or 18.5 MBq (500 µCi) of [67Cu]Cu-SARTATE at either 2 or 4 weeks after tumor cell inoculation. Results PET imaging and ex vivo biodistribution confirmed tumor uptake of [64Cu]Cu-SARTATE and rapid clearance from other tissues. The major clearance tissues were the kidneys (15.6 ± 5.8% IA/g at 24 h post-injection, 11.5 ± 2.8% IA/g at 48 h, n = 3/4). Autoradiography and histological analysis confirmed [64Cu]Cu-SARTATE uptake in viable, SSTR2-positive tumor regions with mean tumor uptakes of 14.1–25.0% IA/g at 24 h. [67Cu]Cu-SARTATE therapy was effective when started 2 weeks after tumor cell inoculation, extending survival by an average of 13 days (30%) compared with the untreated group (mean survival of control group 43.0 ± 8.1 days vs. 55.6 ± 9.1 days for the treated group; p = 0.012). No significant therapeutic effect was observed when [67Cu]Cu-SARTATE was started 4 weeks after tumor cell inoculation, when the tumors would have been larger (control group 14.6 ± 8.5 days; 9.25 MBq group 9.5 ± 1.6 days; 18.5 MBq group 15.6 ± 4.1 days; p = 0.064). Conclusions Clinical experiences of peptide-receptor radionuclide therapy for metastatic disease have been encouraging. This study demonstrates the potential for a theranostic approach using [64/67Cu]Cu-SARTATE for the detection and treatment of SSTR2-positive neuroblastoma MRD.
Collapse
Affiliation(s)
- Jason L J Dearling
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA. .,Harvard Medical School, Boston, MA, 02115, USA.
| | - Ellen M van Dam
- Clarity Pharmaceuticals Ltd., 4 Cornwallis St., Sydney, NSW, 2015, Australia
| | - Matthew J Harris
- Clarity Pharmaceuticals Ltd., 4 Cornwallis St., Sydney, NSW, 2015, Australia
| | - Alan B Packard
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
23
|
Hicks RJ, Roselt PJ, Kallur KG, Tothill RW, Mileshkin L. FAPI PET/CT: Will It End the Hegemony of 18F-FDG in Oncology? J Nucl Med 2020; 62:296-302. [PMID: 33277397 DOI: 10.2967/jnumed.120.256271] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
For over 40 years, 18F-FDG has been the dominant PET tracer in neurology, cardiology, inflammatory diseases, and, most particularly, oncology. Combined with the ability to perform whole-body scanning, 18F-FDG has revolutionized the evaluation of cancer and has stifled the adoption of other tracers, except in situations where low avidity or high background activity limits diagnostic performance. The strength of 18F-FDG has generally been its ability to detect disease in the absence of structural abnormality, thereby enhancing diagnostic sensitivity, but its simultaneous weakness has been a lack of specificity due to diverse pathologies with enhanced glycolysis. Radiotracers that leverage other hallmarks of cancer or specific cell-surface targets are gradually finding a niche in the diagnostic armamentarium. However, none have had sufficient sensitivity to realistically compete with 18F-FDG for evaluation of the broad spectrum of malignancies. Perhaps, this situation is about to change with development of a class of tracers targeting fibroblast activation protein that have low uptake in almost all normal tissues but high uptake in most cancer types. In this review, the development and exciting preliminary clinical data relating to various fibroblast activation protein-specific small-molecule inhibitor tracers in oncology will be discussed along with potential nononcologic applications.
Collapse
Affiliation(s)
- Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Roselt
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Richard W Tothill
- Department of Clinical Pathology and Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Linda Mileshkin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Abstract
Over the past decade, theranostic imaging has emerged as a powerful clinical tool in oncology for identifying patients likely to respond to targeted therapies and for monitoring the response of patients to treatment. Herein, we report a theranostic approach to pretargeted radioimmunotherapy (PRIT) based on a pair of radioisotopes of copper: positron-emitting copper-64 (64Cu, t 1/2 = 12.7 h) and beta particle-emitting copper-67 (67Cu, t 1/2 = 61.8 h). This strategy is predicated on the in vivo ligation between a trans-cyclooctene (TCO)-bearing antibody and a tetrazine (Tz)-based radioligand via the rapid and bioorthogonal inverse electron-demand Diels-Alder reaction. Longitudinal therapy studies were conducted in a murine model of human colorectal carcinoma using an immunoconjugate of the huA33 antibody modified with TCO (huA33-TCO) and a 67Cu-labeled Tz radioligand ([67Cu]Cu-MeCOSar-Tz). The injection of huA33-TCO followed 72 h later by the administration of 18.5, 37.0, or 55.5 MBq of [67Cu]Cu-MeCOSar-Tz produced a dose-dependent therapeutic response, with the median survival time increasing from 68 d for the lowest dose to >200 d for the highest. Furthermore, we observed that mice that received the highest dose of [67Cu]Cu-MeCOSar-Tz in a fractionated manner exhibited improved hematological values without sacrificing therapeutic efficacy. Dual radionuclide experiments in which a single administration of huA33-TCO was followed by separate injections of [64Cu]Cu-MeCOSar-Tz and [67Cu]Cu-MeCOSar-Tz revealed that the positron emission tomography images produced by the former accurately predicted the efficacy of the latter. In these experiments, a correlation was observed between the tumoral uptake of [64Cu]Cu-MeCOSar-Tz and the subsequent therapeutic response to [67Cu]Cu-MeCOSar-Tz.
Collapse
|
25
|
Bolzati C, Duatti A. The emerging value of 64Cu for molecular imaging and therapy. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:329-337. [PMID: 33026210 DOI: 10.23736/s1824-4785.20.03292-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Along with other novel metallic radionuclides, copper-64 (64Cu) is currently being investigated as an alternative option to the gallium-68 (68Ga) and lutetium-177 (177Lu) radiopharmaceuticals widely used for targeting somatostatin receptors, expressed by neuroendocrine tumors (NETs), and recently prostate specific membrane antigen (PSMA), expressed by prostate cancer cells. This interest is mostly driven by the peculiar nuclear properties of 64Cu that make it an almost ideal example of theranostic radionuclide. In fact, 64Cu emits both low-energy positrons, β- particles and a swarm of Auger electrons. This combination of different emissions may allow to collect high-resolution PET images, but also to use the same radiopharmaceutical for eliciting a therapeutic effect. Another unique behavior of 64Cu originates from the fundamental biological role played in organisms by the ionic forms of the copper element, which is naturally involved in a multitude of cellular processes including cell replication. These intrinsic biological characteristics has led to the discovery that 64Cu, under its simplest dicationic form Cu2+, is able to specifically target a variety of cancerous cells and to detect the onset of a metastatic process in its initial stage. This short review reports an outline of the status of 64Cu radiopharmaceuticals and of the most relevant results that are constantly disclosed by preclinical and investigational clinical studies.
Collapse
Affiliation(s)
| | - Adriano Duatti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy -
| |
Collapse
|
26
|
Jalilian AR, Osso JA, Vera-Araujo J, Kumar V, Harris MJ, Gutfilen B, Guérin B, Li H, Zhuravlev F, Chakravarty R, Alirezapour B, Ávila-Rodríguez MA, Khan IU, Aljammaz I, Assaad T, Luurtsema G, Smith J, Duatti A. IAEA contribution to the development of 64Cu radiopharmaceuticals for theranostic applications. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:338-345. [PMID: 33026211 DOI: 10.23736/s1824-4785.20.03302-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Copper-64 is a very attractive radioisotope with unique nuclear properties that allow using it as both a diagnostic and therapeutic agent, thus providing an almost ideal example of a theranostic radionuclide. A characteristic of Cu-64 stems from the intrinsic biological nature of copper ions that play a fundamental role in a large number of cellular processes. Cu-64 is a radionuclide that reflects the natural biochemical pathways of Cu-64 ions, therefore, can be exploited for the detection and therapy of certain malignancies and metabolic diseases. Beside these applications of Cu-64 ions, this radionuclide can be also used for radiolabelling bifunctional chelators carrying a variety of pharmacophores for targeting different biological substrates. These include peptide-based substrates and immunoconjugates as well as small-molecule bioactive moieties. Fueled by the growing interest of Member States (MS) belonging to the International Atomic Energy Agency (IAEA) community, a dedicated Coordinated Research Project (CRP) was initiated in 2016, which recruited thirteen participating MS from four continents. Research activities and collaborations between the participating countries allowed for collection of an impressive series of results, particularly on the production, preclinical evaluation and, in a few cases, clinical evaluation of various 64Cu-radiopharmaceuticals that may have potential impact on future development of the field. Since this CRP was finalized at the beginning of 2020, this short review summarizes outcomes, outputs and results of this project with the purpose to propagate to other MS and to the whole scientific community, some of the most recent achievements on this novel class of theranostic 64Cu-pharmaceuticals.
Collapse
Affiliation(s)
- Amir R Jalilian
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna International Center, Vienna, Austria -
| | - Joao A Osso
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna International Center, Vienna, Austria
| | - Julia Vera-Araujo
- Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna International Center, Vienna, Austria
| | - Vijay Kumar
- Westmead Hospital, Westmead, Sydney, Australia
| | | | - Bianca Gutfilen
- Department of Radiology, Federal University of Rio de Janeiro, Laboratório de Marcação de Células e Moléculas (LMCM), Rio de Janeiro, Brazil
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Centre de Recherche du CHUS (CRCHUS), Centre d'Excellence en Imagerie Médicale (CIMUS), University of Sherbrooke, Sherbrooke, Canada
| | - Hongyu Li
- China Isotope and Radiation Corporation, Beijing, China
| | - Fedor Zhuravlev
- Hevesy Laboratory, Technical University of Denmark (DTU HEALTH TECH), Roskilde, Denmark
| | - Rubel Chakravarty
- Division of Radiopharmaceuticals, Bhabha Atomic Research Center, Mumbai, India
| | - Behrouz Alirezapour
- Radiation Applications Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, Iran
| | - Miguel A Ávila-Rodríguez
- Unit of Cyclotron and Radiopharmaceuticals, Division of Investigation, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Irfan U Khan
- Division of Cyclotron and Allied Radiopharmaceuticals, Institute of Nuclear Medicine and Oncology (INMOL), Lahore, Pakistan
| | - Ibrahim Aljammaz
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Thaer Assaad
- Department of Radioisotope, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Jeff Smith
- MU School of Medicine, University of Missouri, Columbia, MO, USA
| | - Adriano Duatti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
27
|
Eychenne R, Bouvry C, Bourgeois M, Loyer P, Benoist E, Lepareur N. Overview of Radiolabeled Somatostatin Analogs for Cancer Imaging and Therapy. Molecules 2020; 25:4012. [PMID: 32887456 PMCID: PMC7504749 DOI: 10.3390/molecules25174012] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Identified in 1973, somatostatin (SST) is a cyclic hormone peptide with a short biological half-life. Somatostatin receptors (SSTRs) are widely expressed in the whole body, with five subtypes described. The interaction between SST and its receptors leads to the internalization of the ligand-receptor complex and triggers different cellular signaling pathways. Interestingly, the expression of SSTRs is significantly enhanced in many solid tumors, especially gastro-entero-pancreatic neuroendocrine tumors (GEP-NET). Thus, somatostatin analogs (SSAs) have been developed to improve the stability of the endogenous ligand and so extend its half-life. Radiolabeled analogs have been developed with several radioelements such as indium-111, technetium-99 m, and recently gallium-68, fluorine-18, and copper-64, to visualize the distribution of receptor overexpression in tumors. Internal metabolic radiotherapy is also used as a therapeutic strategy (e.g., using yttrium-90, lutetium-177, and actinium-225). With some radiopharmaceuticals now used in clinical practice, somatostatin analogs developed for imaging and therapy are an example of the concept of personalized medicine with a theranostic approach. Here, we review the development of these analogs, from the well-established and authorized ones to the most recently developed radiotracers, which have better pharmacokinetic properties and demonstrate increased efficacy and safety, as well as the search for new clinical indications.
Collapse
Affiliation(s)
- Romain Eychenne
- UPS, CNRS, SPCMIB (Laboratoire de Synthèse et Physico-Chimie de Molécules d’Intérêt Biologique)—UMR 5068, Université de Toulouse, F-31062 Toulouse, France; (R.E.); (E.B.)
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint Herblain, France;
- CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Inserm, Université de Nantes, F-44000 Nantes, France
| | - Christelle Bouvry
- Comprehensive Cancer Center Eugène Marquis, Rennes, F-35000, France;
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes)—UMR 6226, Univ Rennes, F-35000 Rennes, France
| | - Mickael Bourgeois
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint Herblain, France;
- CNRS, CRCINA (Centre de Recherche en Cancérologie et Immunologie Nantes—Angers)—UMR 1232, ERL 6001, Inserm, Université de Nantes, F-44000 Nantes, France
| | - Pascal Loyer
- INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)—UMR_A 1341, UMR_S 1241, Inserm, Univ Rennes, F-35000 Rennes, France;
| | - Eric Benoist
- UPS, CNRS, SPCMIB (Laboratoire de Synthèse et Physico-Chimie de Molécules d’Intérêt Biologique)—UMR 5068, Université de Toulouse, F-31062 Toulouse, France; (R.E.); (E.B.)
| | - Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, F-35000, France;
- INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)—UMR_A 1341, UMR_S 1241, Inserm, Univ Rennes, F-35000 Rennes, France;
| |
Collapse
|
28
|
Houston Z, Bunt J, Chen KS, Puttick S, Howard CB, Fletcher NL, Fuchs AV, Cui J, Ju Y, Cowin G, Song X, Boyd AW, Mahler SM, Richards LJ, Caruso F, Thurecht KJ. Understanding the Uptake of Nanomedicines at Different Stages of Brain Cancer Using a Modular Nanocarrier Platform and Precision Bispecific Antibodies. ACS CENTRAL SCIENCE 2020; 6:727-738. [PMID: 32490189 PMCID: PMC7256936 DOI: 10.1021/acscentsci.9b01299] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Indexed: 06/11/2023]
Abstract
Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.
Collapse
Affiliation(s)
- Zachary
H. Houston
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jens Bunt
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kok-Siong Chen
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- Brigham
and Women’s Hospital, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Simon Puttick
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Commonwealth
Scientific and Industrial Research Organisation, Probing Biosystems
Future Science Platform, Royal Brisbane
and Women’s Hospital, Brisbane, Queensland 4029, Australia
| | - Christopher B. Howard
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L. Fletcher
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Adrian V. Fuchs
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jiwei Cui
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Key
Laboratory of Colloid and Interface Chemistry of the Ministry of Education,
School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yi Ju
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gary Cowin
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Xin Song
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Andrew W. Boyd
- Leukaemia
Foundation Laboratory, QIMR-Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
- Department
of Medicine, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stephen M. Mahler
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Linda J. Richards
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- The
School of Biomedical Sciences, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Frank Caruso
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J. Thurecht
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
29
|
Cullinane C, Jeffery CM, Roselt PD, van Dam EM, Jackson S, Kuan K, Jackson P, Binns D, van Zuylekom J, Harris MJ, Hicks RJ, Donnelly PS. Peptide Receptor Radionuclide Therapy with 67Cu-CuSarTATE Is Highly Efficacious Against a Somatostatin-Positive Neuroendocrine Tumor Model. J Nucl Med 2020; 61:1800-1805. [PMID: 32414949 DOI: 10.2967/jnumed.120.243543] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/16/2020] [Indexed: 01/13/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using radiolabeled octreotate is an effective treatment for somatostatin receptor 2-expressing neuroendocrine tumors. The diagnostic and therapeutic potential of 64Cu and 67Cu, respectively, offers the possibility of using a single somatostatin receptor-targeted peptide conjugate as a theranostic agent. A sarcophagine cage amine ligand, MeCOSar (5-(8-methyl-3,6,10,13,16,19-hexaaza-bicyclo[6.6.6]icosan-1-ylamino)-5-oxopentanoic acid), conjugated to (Tyr3)-octreotate, called 64Cu-CuSarTATE, was demonstrated to be an imaging agent and potential prospective dosimetry tool in 10 patients with neuroendocrine tumors. This study aimed to explore the antitumor efficacy of 67Cu-CuSarTATE in a preclinical model of neuroendocrine tumors and compare it with the standard PRRT agent, 177Lu-LuDOTA-Tyr3-octreotate (177Lu-LuTATE). Methods: The antitumor efficacy of various doses of 67Cu-CuSarTATE in AR42J (rat pancreatic exocrine) tumor-bearing mice was compared with 177Lu-LuTATE. Results: Seven days after a single administration of 67Cu-CuSarTATE (5 MBq), tumor growth was inhibited by 75% compared with vehicle control. Administration of 177Lu-LuTATE (5 MBq) inhibited tumor growth by 89%. Survival was extended from 12 d in the control group to 21 d after treatment with both 67Cu-CuSarTATE and 177Lu-LuTATE. In a second study, the efficacy of fractionated delivery of PRRT was assessed, comparing the efficacy of 30 MBq of 67Cu-CuSarTATE or 177Lu-LuTATE, either as a single intravenous injection or as two 15-MBq fractions 2 wk apart. Treatment of tumors with 2 fractions significantly improved survival over delivery as a single fraction (67Cu-CuSarTATE: 47 vs. 36 d [P = 0.036]; 177Lu-LuTATE: 46 vs. 29 d [P = 0.040]). Conclusion: This study demonstrates that 67Cu-CuSarTATE is well tolerated in BALB/c nude mice and highly efficacious against AR42J tumors in vivo. Administration of 67Cu-CuSarTATE and 177Lu-LuTATE divided into 2 fractions over 2 wk was more efficacious than administration of a single fraction. The antitumor activity of 67Cu-CuSarTATE in the AR42J tumor model demonstrated the suitability of this novel agent for clinical assessment in the treatment of somatostatin receptor 2-expressing neuroendocrine tumors.
Collapse
Affiliation(s)
- Carleen Cullinane
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Peter D Roselt
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ellen M van Dam
- Clarity Pharmaceuticals Ltd., Eveleigh, New South Wales, Australia
| | - Susan Jackson
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kevin Kuan
- Molecular Imaging and Therapy Research Unit, SAHMRI, Adelaide, South Australia, Australia; and
| | - Price Jackson
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - David Binns
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jessica van Zuylekom
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Matthew J Harris
- Clarity Pharmaceuticals Ltd., Eveleigh, New South Wales, Australia
| | - Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.,Centre for Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Kelly JM, Ponnala S, Amor-Coarasa A, Zia NA, Nikolopoulou A, Williams C, Schlyer DJ, DiMagno SG, Donnelly PS, Babich JW. Preclinical Evaluation of a High-Affinity Sarcophagine-Containing PSMA Ligand for 64Cu/ 67Cu-Based Theranostics in Prostate Cancer. Mol Pharm 2020; 17:1954-1962. [PMID: 32286841 DOI: 10.1021/acs.molpharmaceut.0c00060] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The application of small molecules targeting prostate-specific membrane antigen (PSMA) has emerged as a highly promising clinical strategy for visualization and treatment of prostate cancer. Ligands that integrate the ability to both quantify the distribution of radioactivity and treat disease through the use of a matched pair of radionuclides have particular value in clinical and regulatory settings. In this study, we describe the development and preclinical evaluation of RPS-085, a ligand that binds PSMA and serum albumin and exploits the 64/67Cu radionuclide pair for prostate cancer theranostics. RPS-085 was synthesized by conjugation of a PSMA-targeting moiety, an Nε-(2-(4-iodophenyl)acetyl)lysine albumin binding group, and a bifunctionalized MeCOSar chelator. The IC50 of the metal-free RPS-085 was determined in a competitive binding assay. The affinity for human serum albumin of the radiolabeled compound was determined by high-performance affinity chromatography. Radiolabeling was performed in NH4OAc buffer at 25 °C. The stability of the radiolabeled compounds was assessed in vitro and in vivo. The biodistribution of [64/67Cu]Cu-RPS-085 was determined following intravenous administration to male BALB/c mice bearing LNCaP tumor xenografts. The radiochemical yields of [64/67Cu]Cu-RPS-085 were nearly quantitative after 20 min. The metal-free complex is a potent inhibitor of PSMA (IC50 = 29 ± 2 nM), and the radiolabeled compound has moderate affinity for human serum albumin (Kd = 9.9 ± 1.7 μM). Accumulation of the tracer in mice was primarily evident in tumor and kidneys. Activity in all other tissues, including blood, was negligible, and the radiolabeled compounds demonstrated high stability in vitro and in vivo. Tumor activity reached a maximum at 4 h post injection (p.i.) and cleared gradually over a period of 96 h. By contrast, activity in the kidney cleared rapidly from 4 to 24 h p.i. As a consequence, by 24 h p.i., the tumor-to-kidney ratio exceeds 2, and the predicted dose to tumors is significantly greater than the dose to kidneys. [64Cu]Cu-RPS-085 combines rapid tissue distribution and clearance with prolonged retention in LNCaP tumor xenografts. The pharmacokinetics should enable radioligand therapy using [67Cu]Cu-RPS-085. By virtue of its rapid kidney clearance, the therapeutic index of [67Cu]Cu-RPS-085 likely compares favorably to its parent structure, [177Lu]Lu-RPS-063, a highly avid PSMA-targeting compound. On this basis, [64/67Cu]Cu-RPS-085 show great promise as PSMA-targeting theranostic ligands for prostate cancer imaging and therapy.
Collapse
Affiliation(s)
- James M Kelly
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Shashikanth Ponnala
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Alejandro Amor-Coarasa
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Nicholas A Zia
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Anastasia Nikolopoulou
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States.,Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, New York 10021, United States
| | - Clarence Williams
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States
| | - David J Schlyer
- Brookhaven National Laboratory, Upton, New York 11973, United States.,Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Stephen G DiMagno
- College of Pharmacy, University of Illinois-Chicago, Chicago, Illinois 60612, United States
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - John W Babich
- Division of Radiopharmaceutical Sciences and MI3 Institute, Department of Radiology, Weill Cornell Medicine, New York, New York 10021, United States.,Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, New York 10021, United States.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, United States
| |
Collapse
|
31
|
|
32
|
Zia NA, Cullinane C, Van Zuylekom JK, Waldeck K, McInnes LE, Buncic G, Haskali MB, Roselt PD, Hicks RJ, Donnelly PS. A Bivalent Inhibitor of Prostate Specific Membrane Antigen Radiolabeled with Copper‐64 with High Tumor Uptake and Retention. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201908964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Nicholas A. Zia
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Carleen Cullinane
- Sir Peter MacCallum Department of Oncology University of Melbourne Melbourne 3010 Vic. Australia
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | | | - Kelly Waldeck
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Lachlan E. McInnes
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Gojko Buncic
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Mohammad B. Haskali
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Peter D. Roselt
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Rodney J. Hicks
- Sir Peter MacCallum Department of Oncology University of Melbourne Melbourne 3010 Vic. Australia
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Paul S. Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| |
Collapse
|
33
|
Functional and anatomical imaging in pediatric oncology: which is best for which tumors. Pediatr Radiol 2019; 49:1534-1544. [PMID: 31620853 DOI: 10.1007/s00247-019-04489-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/19/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
Functional imaging techniques are playing an increasingly important role in the management of pediatric cancer. Technological advances have pushed the development of hybrid imaging techniques, including positron emission tomography (PET)/CT, PET/MR and single-photon emission computed tomography (SPECT)/CT. Together with an increasing need to identify surrogate biomarkers for response to novel therapies, the use of functional imaging techniques, which had been reserved primarily for lymphoma patients, is now being recognized as standard of care for the management of many other pediatric solid tumors. The purpose of this review is to summarize recent data describing the use of functional and metabolic imaging strategies for the staging and response assessment of common pediatric solid tumors, and to offer some guidance as to which techniques are most appropriate for which tumor types.
Collapse
|
34
|
Zia NA, Cullinane C, Van Zuylekom JK, Waldeck K, McInnes LE, Buncic G, Haskali MB, Roselt PD, Hicks RJ, Donnelly PS. A Bivalent Inhibitor of Prostate Specific Membrane Antigen Radiolabeled with Copper‐64 with High Tumor Uptake and Retention. Angew Chem Int Ed Engl 2019; 58:14991-14994. [DOI: 10.1002/anie.201908964] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Nicholas A. Zia
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Carleen Cullinane
- Sir Peter MacCallum Department of Oncology University of Melbourne Melbourne 3010 Vic. Australia
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | | | - Kelly Waldeck
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Lachlan E. McInnes
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Gojko Buncic
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| | - Mohammad B. Haskali
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Peter D. Roselt
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Rodney J. Hicks
- Sir Peter MacCallum Department of Oncology University of Melbourne Melbourne 3010 Vic. Australia
- Research Division Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
- Centre for Cancer Imaging Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
| | - Paul S. Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute University of Melbourne Melbourne 3010 Vic. Australia
| |
Collapse
|
35
|
|
36
|
Hicks RJ, Jackson P, Kong G, Ware RE, Hofman MS, Pattison DA, Akhurst TA, Drummond E, Roselt P, Callahan J, Price R, Jeffery CM, Hong E, Noonan W, Herschtal A, Hicks LJ, Hedt A, Harris M, Paterson BM, Donnelly PS. 64Cu-SARTATE PET Imaging of Patients with Neuroendocrine Tumors Demonstrates High Tumor Uptake and Retention, Potentially Allowing Prospective Dosimetry for Peptide Receptor Radionuclide Therapy. J Nucl Med 2018; 60:777-785. [PMID: 30442752 DOI: 10.2967/jnumed.118.217745] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/01/2018] [Indexed: 01/08/2023] Open
Abstract
Imaging of somatostatin receptor expression is an established technique for staging of neuroendocrine neoplasia and determining the suitability of patients for peptide receptor radionuclide therapy. PET/CT using 68Ga-labeled somatostatin analogs is superior to earlier agents, but the rapid physical decay of the radionuclide poses logistic and regulatory challenges. 64Cu has attractive physical characteristics for imaging and provides a diagnostic partner for the therapeutic radionuclide 67Cu. Based on promising preclinical studies, we have performed a first-time-in-humans trial of 64Cu-MeCOSar-Tyr3-octreotate (64Cu-SARTATE) to assess its safety and ability to localize disease at early and late imaging time-points. Methods: In a prospective trial, 10 patients with known neuroendocrine neoplasia and positive for uptake on 68Ga-DOTA-octreotate (68Ga-DOTATATE) PET/CT underwent serial PET/CT imaging at 30 min, 1 h, 4 h, and 24 h after injection of 64Cu-SARTATE. Adverse reactions were recorded, and laboratory testing was performed during infusion and at 1 and 7 d after imaging. Images were analyzed for lesion and normal-organ uptake and clearance to assess lesion contrast and perform dosimetry estimates. Results: 64Cu-SARTATE was well tolerated during infusion and throughout the study, with 3 patients experiencing mild infusion-related events. High lesion uptake and retention were observed at all imaging time-points. There was progressive hepatic clearance over time, providing the highest lesion-to-liver contrast at 24 h. Image quality remained high at this time. Comparison of 64Cu-SARTATE PET/CT obtained at 4 h to 68Ga-DOTATATE PET/CT obtained at 1 h indicated comparable or superior lesion detection in all patients, especially in the liver. As expected, the highest early physiologic organ uptake was in the kidneys, liver, and spleen. Conclusion: 64Cu-SARTATE is safe and has excellent imaging characteristics. High late-retention in tumor and clearance from the liver suggest suitability for diagnostic studies and for prospective dosimetry for 67Cu-SARTATE peptide receptor radionuclide therapy, and the half-life of 64Cu would also facilitate good-manufacturing-practice production and distribution to sites without access to 68Ga.
Collapse
Affiliation(s)
- Rodney J Hicks
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Price Jackson
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Grace Kong
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Robert E Ware
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David A Pattison
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Timothy A Akhurst
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Elizabeth Drummond
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Peter Roselt
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jason Callahan
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Roger Price
- Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Washington, Australia
| | - Charmaine M Jeffery
- Medical Technology and Physics, Sir Charles Gairdner Hospital, Nedlands, Washington, Australia
| | - Emily Hong
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Wayne Noonan
- Liverpool Hospital, Liverpool, New South Wales, Australia
| | - Alan Herschtal
- Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Lauren J Hicks
- Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Amos Hedt
- Clarity Pharmaceuticals Ltd., Eveleigh, New South Wales, Australia
| | - Matthew Harris
- Clarity Pharmaceuticals Ltd., Eveleigh, New South Wales, Australia
| | - Brett M Paterson
- School of Chemistry, Monash University, Victoria, Australia; and
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
37
|
David T, Hlinová V, Kubíček V, Bergmann R, Striese F, Berndt N, Szöllősi D, Kovács T, Máthé D, Bachmann M, Pietzsch HJ, Hermann P. Improved Conjugation, 64-Cu Radiolabeling, in Vivo Stability, and Imaging Using Nonprotected Bifunctional Macrocyclic Ligands: Bis(Phosphinate) Cyclam (BPC) Chelators. J Med Chem 2018; 61:8774-8796. [PMID: 30180567 DOI: 10.1021/acs.jmedchem.8b00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bifunctional derivatives of bis(phosphinate)-bearing cyclam (BPC) chelators bearing a carboxylate, amine, isothiocyanate, azide, or cyclooctyne in the BP side chain were synthesized. Conjugations required no protection of phosphinate or ring secondary amine groups. The ring amines were not reactive (proton protected) at pH < ∼8. For isothiocyanate coupling, oligopeptide N-terminal α-amines were more suitable than alkyl amines, e.g., Lys ω-amine (p Ka ∼7.5-8.5 and ∼10-11, respectively) due to lower basicity. The Cu-64 labeling was efficient at room temperature (specific activity ∼100 GBq/μmol; 25 °C, pH 6.2, ∼100 ligand equiv, 10 min). A representative Cu-64-BPC was tested in vivo showing fast clearance and no nonspecific radioactivity deposition. The monoclonal anti-PSCA antibody 7F5 conjugates with thiocyanate BPC derivative or NODAGA were radiolabeled and studied in PC3-PSCA tumor bearing mice by PET. The radiolabeled BPC conjugate was accumulated in the prostate tumor with a low off-target uptake, unlike Cu-64-labeled NODAGA-antibody conjugate. The BPC chelators have a great potential for theranostic applications of the Cu-64/Cu-67 matched pair.
Collapse
Affiliation(s)
- Tomáš David
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic.,Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Veronika Hlinová
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| | - Vojtěch Kubíček
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| | - Ralf Bergmann
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Franziska Striese
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Nicole Berndt
- Partner Site Dresden , German Cancer Consortium (DKTK) , Fetscherstrasse 74 , 01307 Dresden , Germany.,German Cancer Research Center (DKFZ) , Im Neuenheimer Feld 280 , 69120 Heidelberg , Germany
| | - Dávid Szöllősi
- Department of Biophysics and Radiation Biology , Semmelweis University , Tűzoltó utca 37-47 , H-1094 Budapest , Hungary
| | - Tibor Kovács
- Institute of Radiochemistry and Radioecology , University of Pannonia , Egyetem St. 10 , H-8200 Veszprém , Hungary.,Social Organization for Radioecological Cleanliness , P.O. Box 158, H-8200 Veszprém , Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology , Semmelweis University , Tűzoltó utca 37-47 , H-1094 Budapest , Hungary
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany.,Tumor Immunology, University Cancer Center (UCC) , "Carl Gustav Carus" Technische Universität Dresden , Fetscherstrasse 74 , 01307 Dresden , Germany.,National Center for Tumor Diseases (NCT) , "Carl Gustav Carus" Technische Universität Dresden , Fetscherstrasse 74 , 01307 Dresden , Germany
| | - Hans-Jürgen Pietzsch
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Petr Hermann
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| |
Collapse
|
38
|
Ahmedova A, Todorov B, Burdzhiev N, Goze C. Copper radiopharmaceuticals for theranostic applications. Eur J Med Chem 2018; 157:1406-1425. [DOI: 10.1016/j.ejmech.2018.08.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/15/2018] [Accepted: 08/18/2018] [Indexed: 12/12/2022]
|
39
|
The emerging role of copper-64 radiopharmaceuticals as cancer theranostics. Drug Discov Today 2018; 23:1489-1501. [DOI: 10.1016/j.drudis.2018.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/02/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
|
40
|
Geuijen CAW, De Nardis C, Maussang D, Rovers E, Gallenne T, Hendriks LJA, Visser T, Nijhuis R, Logtenberg T, de Kruif J, Gros P, Throsby M. Unbiased Combinatorial Screening Identifies a Bispecific IgG1 that Potently Inhibits HER3 Signaling via HER2-Guided Ligand Blockade. Cancer Cell 2018; 33:922-936.e10. [PMID: 29763625 DOI: 10.1016/j.ccell.2018.04.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 02/26/2018] [Accepted: 04/09/2018] [Indexed: 01/21/2023]
Abstract
HER2-driven cancers require phosphatidylinositide-3 kinase (PI3K)/Akt signaling through HER3 to promote tumor growth and survival. The therapeutic benefit of HER2-targeting agents, which depend on PI3K/Akt inhibition, can be overcome by hyperactivation of the heregulin (HRG)/HER3 pathway. Here we describe an unbiased phenotypic combinatorial screening approach to identify a bispecific immunoglobulin G1 (IgG1) antibody against HER2 and HER3. In tumor models resistant to HER2-targeting agents, the bispecific IgG1 potently inhibits the HRG/HER3 pathway and downstream PI3K/Akt signaling via a "dock & block" mechanism. This bispecific IgG1 is a potentially effective therapy for breast cancer and other tumors with hyperactivated HRG/HER3 signaling.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/pharmacology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Screening Assays, Antitumor
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/pharmacology
- MCF-7 Cells
- Mice
- Models, Molecular
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Binding/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-3/chemistry
- Receptor, ErbB-3/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | - Camilla De Nardis
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | | - Piet Gros
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | | |
Collapse
|
41
|
Rylova SN, Stoykow C, Del Pozzo L, Abiraj K, Tamma ML, Kiefer Y, Fani M, Maecke HR. The somatostatin receptor 2 antagonist 64Cu-NODAGA-JR11 outperforms 64Cu-DOTA-TATE in a mouse xenograft model. PLoS One 2018; 13:e0195802. [PMID: 29668724 PMCID: PMC5906006 DOI: 10.1371/journal.pone.0195802] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/29/2018] [Indexed: 01/29/2023] Open
Abstract
Copper-64 is an attractive radionuclide for PET imaging and is frequently used in clinical applications. The aim of this study was to perform a side-by-side comparison of the in vitro and in vivo performance of 64Cu-NODAGA-JR11 (NODAGA = 1,4,7-triazacyclononane,1-glutaric acid,4,7-acetic acid, JR11 = p-Cl-Phe-cyclo(D-Cys-Aph(Hor)-D-Aph(cbm)-Lys-Thr-Cys)D-Tyr-NH2), a somatostatin receptor 2 antagonist, with the clinically used sst2 agonist 64Cu-DOTA-TATE ((TATE = D-Phe-cyclo(Cys-Tyr-D-Trp-Lys-Thr-Cys)Thr). In vitro studies demonstrated Kd values of 5.7±0.95 nM (Bmax = 4.1±0.18 nM) for the antagonist 64/natCu-NODAGA-JR11 and 20.1±4.4. nM (Bmax = 0.48±0.18 nM) for the agonist 64/natCu-DOTA-TATE. Cell uptake studies showed the expected differences between agonists and antagonists. Whereas 64Cu-DOTA-TATE (the agonist) showed very effective internalization in the cell culture assay (with 50% internalized at 4 hours post-peptide addition under the given experimental conditions), 64Cu-NODAGA-JR11 (the antagonist) showed little internalization but strong receptor-mediated uptake at the cell membrane. Biodistribution studies of 64Cu-NODAGA-JR11 showed rapid blood clearance and tumor uptake with increasing tumor-to-relevant organ ratios within the first 4 hours and in some cases, 24 hours, respectively. The tumor washout was slow or non-existent in the first 4 hours, whereas the kidney washout was very efficient, leading to high and increasing tumor-to-kidney ratios over time. Specificity of tumor uptake was proven by co-injection of high excess of non-radiolabeled peptide, which led to >80% tumor blocking. 64Cu-DOTA-TATE showed less favorable pharmacokinetics, with the exception of lower kidney uptake. Blood clearance was distinctly slower and persistent higher blood values were found at 24 hours. Uptake in the liver and lung was relatively high and also persistent. The tumor uptake was specific and similar to that of 64Cu-NODAGA-JR11 at 1 h, but release from the tumor was very fast, particularly between 4 and 24 hours. Tumor-to-normal organ ratios were distinctly lower after 1 hour. This is indicative of insufficient in vivo stability. PET studies of 64Cu-NODAGA-JR11 reflected the biodistribution data with nicely delineated tumor and low background. 64Cu-NODAGA-JR11 shows promising pharmacokinetic properties for further translation into the clinic.
Collapse
Affiliation(s)
- Svetlana N. Rylova
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Stoykow
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Luigi Del Pozzo
- Division of Radiological Chemistry, University Hospital Basel, Basel, Switzerland
| | - Keelara Abiraj
- Division of Radiological Chemistry, University Hospital Basel, Basel, Switzerland
| | - Maria Luisa Tamma
- Division of Radiological Chemistry, University Hospital Basel, Basel, Switzerland
| | - Yvonne Kiefer
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melpomeni Fani
- Division of Radiological Chemistry, University Hospital Basel, Basel, Switzerland
| | - Helmut R. Maecke
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
42
|
Paúrová M, David T, Císařová I, Lubal P, Hermann P, Kotek J. Optimization of the selectivity and rate of copper radioisotope complexation: formation and dissociation kinetic studies of 1,4,8-trimethylcyclam-based ligands with different coordinating pendant arms. NEW J CHEM 2018. [DOI: 10.1039/c8nj00419f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Influence of coordinating pendant arm character on selectivity and rate of copper(ii) complexation was investigated to optimize ligands for radiomedicinal use.
Collapse
Affiliation(s)
- Monika Paúrová
- Department of Inorganic Chemistry
- Faculty of Science
- Charles University
- Czech Republic
| | - Tomáš David
- Department of Inorganic Chemistry
- Faculty of Science
- Charles University
- Czech Republic
| | - Ivana Císařová
- Department of Inorganic Chemistry
- Faculty of Science
- Charles University
- Czech Republic
| | - Přemysl Lubal
- Department of Chemistry
- Faculty of Science
- Masaryk University
- Brno
- Czech Republic
| | - Petr Hermann
- Department of Inorganic Chemistry
- Faculty of Science
- Charles University
- Czech Republic
| | - Jan Kotek
- Department of Inorganic Chemistry
- Faculty of Science
- Charles University
- Czech Republic
| |
Collapse
|
43
|
McInnes LE, Rudd SE, Donnelly PS. Copper, gallium and zirconium positron emission tomography imaging agents: The importance of metal ion speciation. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2017.05.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
Jamier V, Mume E, Papamicaël C, Smith SV. Exploring the synthesis and metal complexation behavior of mono and bis substituted hexaazamacrocyclic cage derivatives. INORG CHEM COMMUN 2017. [DOI: 10.1016/j.inoche.2017.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Yap ML, McFadyen JD, Wang X, Zia NA, Hohmann JD, Ziegler M, Yao Y, Pham A, Harris M, Donnelly PS, Hogarth PM, Pietersz GA, Lim B, Peter K. Targeting Activated Platelets: A Unique and Potentially Universal Approach for Cancer Imaging. Am J Cancer Res 2017; 7:2565-2574. [PMID: 28819447 PMCID: PMC5558553 DOI: 10.7150/thno.19900] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/26/2017] [Indexed: 12/27/2022] Open
Abstract
Rationale The early detection of primary tumours and metastatic disease is vital for successful therapy and is contingent upon highly specific molecular markers and sensitive, non-invasive imaging techniques. We hypothesized that the accumulation of activated platelets within tumours is a general phenomenon and thus represents a novel means for the molecular imaging of cancer. Here we investigate a unique single chain antibody (scFv), which specifically targets activated platelets, as a novel biotechnological tool for molecular imaging of cancer. Methods The scFvGPIIb/IIIa, which binds specifically to the activated form of the platelet integrin receptor GPIIb/IIIa present on activated platelets, was conjugated to either Cy7, 64Cu or ultrasound-enhancing microbubbles. Using the Cy7 labelled scFvGPIIb/IIIa, fluorescence imaging was performed in mice bearing four different human tumour xenograft models; SKBr3, MDA-MB-231, Ramos and HT-1080 cells. Molecular imaging via PET and ultrasound was performed using the scFvGPIIb/IIIa-64Cu and scFvGPIIb/IIIa-microbubbles, respectively, to further confirm specific targeting of scFvGPIIb/IIIa to activated platelets in the tumour stroma. Results Using scFvGPIIb/IIIa we successfully showed specific targeting of activated platelets within the microenvironment of human tumour xenografts models via three different molecular imaging modalities. The presence of platelets within the tumour microenvironment, and as such their relevance as a molecular target epitope in cancer was further confirmed via immunofluorescence of human tumour sections of various cancer types, thus validating the translational importance of our novel approach to human disease. Conclusion Our study provides proof of concept for imaging and localization of tumours by molecular targeting activated platelets. We illustrate the utility of a unique scFv as a versatile biotechnological tool which can be conjugated to various contrast agents for molecular imaging of cancer using three different imaging modalities. These findings warrant further development of this activated platelet specific scFvGPIIb/IIIa, potentially as a universal marker for cancer diagnosis and ultimately for drug delivery in an innovative theranostic approach.
Collapse
|
46
|
Marciniak A, Brasuń J. Somatostatin analogues labeled with copper radioisotopes: current status. J Radioanal Nucl Chem 2017; 313:279-289. [PMID: 28804185 PMCID: PMC5533839 DOI: 10.1007/s10967-017-5323-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Indexed: 12/23/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) is a promising way to treat patients with inoperable tumors or metastatic neuroendocrine tumors. This therapeutic strategy is using radiolabeled peptides, which are capable of selective biding to receptors overexpressed in the cancer cells. One of the group of receptor-avid peptide used in the PRRT are the analogues of somatostatin (SST) connected to the complexes of radionuclides (e.g. 90Y, 177Lu or 111In). Many studies have shown that radiopharmaceuticals based on Cu radioisotopes are promising for the diagnosis and treatment of various cancers. This mini-review focuses on recent developments and summarises the results of multiple studies addressing SST agonists and antagonists radiolabeled to Cu radioisotopes.
Collapse
Affiliation(s)
- Aleksandra Marciniak
- Department of Inorganic Chemistry, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Justyna Brasuń
- Department of Inorganic Chemistry, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
47
|
Imberti C, Terry SYA, Cullinane C, Clarke F, Cornish GH, Ramakrishnan NK, Roselt P, Cope AP, Hicks RJ, Blower PJ, Ma MT. Enhancing PET Signal at Target Tissue in Vivo: Dendritic and Multimeric Tris(hydroxypyridinone) Conjugates for Molecular Imaging of α vβ 3 Integrin Expression with Gallium-68. Bioconjug Chem 2017; 28:481-495. [PMID: 27966893 PMCID: PMC5314429 DOI: 10.1021/acs.bioconjchem.6b00621] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 11/23/2016] [Indexed: 12/12/2022]
Abstract
Tris(hydroxypyridinone) chelators conjugated to peptides can rapidly complex the positron-emitting isotope gallium-68 (68Ga) under mild conditions, and the resulting radiotracers can delineate peptide receptor expression at sites of diseased tissue in vivo. We have synthesized a dendritic bifunctional chelator containing nine 1,6-dimethyl-3-hydroxypyridin-4-one groups (SCN-HP9) that can coordinate up to three Ga3+ ions. This derivative has been conjugated to a trimeric peptide (RGD3) containing three peptide groups that target the αvβ3 integrin receptor. The resulting dendritic compound, HP9-RGD3, can be radiolabeled in 97% radiochemical yield at a 3-fold higher specific activity than its homologues HP3-RGD and HP3-RGD3 that contain only a single metal binding site. PET scanning and biodistribution studies show that [68Ga(HP9-RGD3)] demonstrates higher receptor-mediated tumor uptake in animals bearing U87MG tumors that overexpress αvβ3 integrin than [68Ga(HP3-RGD)] and [68Ga(HP3-RGD3)]. However, concomitant nontarget organ retention of [68Ga(HP9-RGD3)] results in low tumor to nontarget organ contrast in PET images. On the other hand, the trimeric peptide homologue containing a single tris(hydroxypyridinone) chelator, [68Ga(HP3-RGD3)], clears nontarget organs and exhibits receptor-mediated uptake in mice bearing tumors and in mice with induced rheumatoid arthritis. PET imaging with [68Ga(HP3-RGD3)] enables clear delineation of αvβ3 integrin receptor expression in vivo.
Collapse
Affiliation(s)
- Cinzia Imberti
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Samantha Y. A. Terry
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Carleen Cullinane
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Fiona Clarke
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Georgina H. Cornish
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Nisha K. Ramakrishnan
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Peter Roselt
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Andrew P. Cope
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Rodney J. Hicks
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Philip J. Blower
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Michelle T. Ma
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
48
|
Ziegler M, Alt K, Paterson BM, Kanellakis P, Bobik A, Donnelly PS, Hagemeyer CE, Peter K. Highly Sensitive Detection of Minimal Cardiac Ischemia using Positron Emission Tomography Imaging of Activated Platelets. Sci Rep 2016; 6:38161. [PMID: 27909290 PMCID: PMC5133579 DOI: 10.1038/srep38161] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/04/2016] [Indexed: 01/12/2023] Open
Abstract
A reliable method for the diagnosis of minimal cardiac ischemia would meet a strong demand for the sensitive diagnosis of coronary artery disease in cardiac stress testing and risk stratification in patients with chest pain but unremarkable ECGs and biomarkers. We hypothesized that platelets accumulate early on in ischemic myocardium and a newly developed technology of non-invasive molecular PET imaging of activated platelets can thus detect minimal degrees of myocardial ischemia. To induce different degrees of minimal cardiac ischemia, the left anterior descending artery (LAD) was ligated for 10, 20 or 60 min. Mice were injected with a newly generated scFvanti-GPIIb/IIIa-64CuMeCOSar radiotracer, composed of a single-chain antibody that only binds to activated integrin GPIIb/IIIa (αIIbβIII) and thus to activated platelets, and a sarcophagine cage MeCOSar complexing the long half-life PET tracer copper-64. A single PET/CT scan was performed. Evans Blue/TTC staining to detect necrosis as well as classical serological biomarkers like Troponin I and heart-type fatty acid-binding protein (H-FABP) were negative, whereas PET imaging of activated platelets was able to detect small degrees of ischemia. Taken together, molecular PET imaging of activated platelets represents a unique and highly sensitive method to detect minimal cardiac ischemia.
Collapse
Affiliation(s)
- Melanie Ziegler
- Atherothrombosis and Vascular Biology, Baker IDI Heart &Diabetes Institute, Melbourne, Australia
| | - Karen Alt
- Atherothrombosis and Vascular Biology, Baker IDI Heart &Diabetes Institute, Melbourne, Australia.,Vascular Biotechnology, Baker IDI Heart &Diabetes Institute, Melbourne, Australia
| | - Brett M Paterson
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Peter Kanellakis
- Vascular Biology &Atherosclerosis, Baker IDI Heart &Diabetes Institute, Melbourne, Australia
| | - Alex Bobik
- Vascular Biology &Atherosclerosis, Baker IDI Heart &Diabetes Institute, Melbourne, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Christoph E Hagemeyer
- Vascular Biotechnology, Baker IDI Heart &Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker IDI Heart &Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia.,RMIT University, Melbourne, Australia
| |
Collapse
|
49
|
Template synthesis and X-ray structure of the tris-glyoximate iron(II) clathrochelates with terminal reactive groups. Inorganica Chim Acta 2016. [DOI: 10.1016/j.ica.2016.08.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
50
|
Charron CL, Farnsworth AL, Roselt PD, Hicks RJ, Hutton CA. Recent developments in radiolabelled peptides for PET imaging of cancer. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.07.083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|