1
|
Najdian A, Beiki D, Abbasi M, Gholamrezanezhad A, Ahmadzadehfar H, Amani AM, Ardestani MS, Assadi M. Exploring innovative strides in radiolabeled nanoparticle progress for multimodality cancer imaging and theranostic applications. Cancer Imaging 2024; 24:127. [PMID: 39304961 DOI: 10.1186/s40644-024-00762-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
Multimodal imaging unfolds as an innovative approach that synergistically employs a spectrum of imaging techniques either simultaneously or sequentially. The integration of computed tomography (CT), magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), positron emission tomography (PET), and optical imaging (OI) results in a comprehensive and complementary understanding of complex biological processes. This innovative approach combines the strengths of each method and overcoming their individual limitations. By harmoniously blending data from these modalities, it significantly improves the accuracy of cancer diagnosis and aids in treatment decision-making processes. Nanoparticles possess a high potential for facile functionalization with radioactive isotopes and a wide array of contrast agents. This strategic modification serves to augment signal amplification, significantly enhance image sensitivity, and elevate contrast indices. Such tailored nanoparticles constructs exhibit a promising avenue for advancing imaging modalities in both preclinical and clinical setting. Furthermore, nanoparticles function as a unified nanoplatform for the co-localization of imaging agents and therapeutic payloads, thereby optimizing the efficiency of cancer management strategies. Consequently, radiolabeled nanoparticles exhibit substantial potential in driving forward the realms of multimodal imaging and theranostic applications. This review discusses the potential applications of molecular imaging in cancer diagnosis, the utilization of nanotechnology-based radiolabeled materials in multimodal imaging and theranostic applications, as well as recent advancements in this field. It also highlights challenges including cytotoxicity and regulatory compliance, essential considerations for effective clinical translation of nanoradiopharmaceuticals in multimodal imaging and theranostic applications.
Collapse
Affiliation(s)
- Atena Najdian
- The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Davood Beiki
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California (USC), 1441 Eastlake Ave Ste 2315, Los Angeles, CA, 90089, USA
| | - Hojjat Ahmadzadehfar
- Department of Nuclear Medicine, Klinikum Westfalen, Dortmund, Germany
- Department of Nuclear Medicine, Institute of Radiology, Neuroradiology and Nuclear Medicine, University Hospital Knappschaftskrankenhaus, Bochum, Germany
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
2
|
Basuli F, Vasalatiy O, Shi J, Lane KC, Escorcia FE, Swenson RE. Preparation of a Zirconium-89 Labeled Clickable DOTA Complex and Its Antibody Conjugate. Pharmaceuticals (Basel) 2024; 17:480. [PMID: 38675440 PMCID: PMC11053460 DOI: 10.3390/ph17040480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Desferrioxamine B (DFO) is the clinical standard chelator for preparing zirconium-89 labeled antibodies. In the current study, the stabilities of a zirconium-89 labeled panitumumab (PAN; Vectibix®) with three different chelators (DFO, DFO*, and DOTA) were compared. PAN is an anti-HER1/EGFR monoclonal antibody approved by the FDA for the treatment of HER1-expressing colorectal cancers and was used as the model antibody for this study. DFO/DFO* conjugates of PAN were directly radiolabeled with zirconium-89 at room temperature to produce [89Zr]Zr-DFO/DFO*-PAN conjugates following a well-established procedure. A zirconium-89 labeled DOTA-PAN conjugate was prepared by an indirect radiolabeling method. A cyclooctyne-linked DOTA chelator (BCN-DOTA-GA) was first radiolabeled with zirconium-89 at 90 °C under a two-step basic pH adjustment method followed by conjugation with PAN-tetrazene at 37 °C to produce a labeled conjugate, BCN-[89Zr]Zr-DOTA-GA-PAN. High reproducibility of the radiolabeling was observed via this two-step basic pH adjustment. The overall radiochemical yield was 40-50% (n = 12, decay uncorrected) with a radiochemical purity of >95% in 2 h synthesis time. All three conjugates were stable in whole human serum for up to 7 days at 37 °C. The kinetic inertness of the conjugates was assessed against the EDTA challenge. BCN-[89Zr]Zr-DOTA-GA-PAN exhibited excellent inertness followed by [89Zr]Zr-DFO*-PAN. [89Zr]Zr-DFO-PAN displayed the lowest level of inertness.
Collapse
Affiliation(s)
- Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (O.V.); (J.S.); (K.C.L.); (R.E.S.)
| | - Olga Vasalatiy
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (O.V.); (J.S.); (K.C.L.); (R.E.S.)
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (O.V.); (J.S.); (K.C.L.); (R.E.S.)
| | - Kelly C. Lane
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (O.V.); (J.S.); (K.C.L.); (R.E.S.)
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (O.V.); (J.S.); (K.C.L.); (R.E.S.)
| |
Collapse
|
3
|
Deiser S, Drexler M, Moreno-Alcántar G, Irl M, Schmidt C, Günther T, Casini A. Synthesis of 177Lu-Labeled, Somatostatin-2 Receptor-Targeted Metalla-Assemblies: Challenges in the Design of Supramolecular Radiotherapeutics. Inorg Chem 2023; 62:20710-20720. [PMID: 37556427 DOI: 10.1021/acs.inorgchem.3c02090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Self-assembled supramolecular coordination complexes (SCCs) hold promise for biomedical applications in cancer therapy, although their potential in the field of nuclear medicine is still substantially unexplored. Therefore, in this study an exo-functionalized cationic [Pd2L2]4+ metallacycle (L = 3,5-bis(3-ethynylpyridine)phenyl), targeted to the somatostatin-2 receptor (sst2R) and featuring the DOTA chelator (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) in order to bind the β-- and γ-emitter lutetium-177, was synthesized by self-assembly following ligand synthesis via standard solid-phase peptide synthesis (SPPS). This metallacycle was then characterized by reverse-phase high-performance liquid chromatography (RP-HPLC), electrospray ionization mass spectrometry (ESI-MS), and 1H and 1H-DOSY NMR (DOSY = diffusion-ordered spectroscopy). A procedure for the radiolabeling of the metallacycle with 177Lu was also optimized. The resulting [nat/177Lu]Lu-DOTA-metallacycle, termed [nat/177Lu]Lu-Cy, was evaluated concerning its stability and in vitro properties. The compound was more lipophilic compared to the reference [177Lu]Lu-DOTA-TATE (logPOct/H2O = -0.85 ± 0.10 versus -3.67 ± 0.04, respectively). While [natLu]Lu-Cy revealed low stability in a DMEM/F12 GlutaMax medium, it demonstrated good stability in other aqueous media as well as in DMSO. A high sst2R binding affinity (expressed as IC50) was determined in CHOsst2 cells (Chinese hamster ovary cells that were stably transfected with human sst2R). Moreover, the metallacycle exhibited high human serum albumin binding, as assessed by high-performance affinity chromatography (HPAC), and moderate stability in human serum compared to [177Lu]Lu-DOTA-TATE (TATE = (Tyr3)-octreotate). In order to improve stability, a heteroleptic approach was used to develop a less sterically hindered cage-like SCC that is potentially endowed with host-guest chemistry capability, which has been preliminarily characterized by RP-HPLC and ESI-MS. Overall, our initial results encourage future studies on sst2R-directed SCCs and have led to new insights into the chemistry of ss2R-directed SCCs for radiopharmaceutical applications.
Collapse
Affiliation(s)
- Sandra Deiser
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Marike Drexler
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Maximilian Irl
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
| | - Claudia Schmidt
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Thomas Günther
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Walther-Meißner-Str. 3, 85748 Garching b. München, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| |
Collapse
|
4
|
Relevance of Volumetric Parameters Applied to [ 68Ga]Ga-DOTATOC PET/CT in NET Patients Treated with PRRT. Diagnostics (Basel) 2023; 13:diagnostics13040606. [PMID: 36832093 PMCID: PMC9955025 DOI: 10.3390/diagnostics13040606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND this study aims to explore the prognostic and predictive role of volumetric parameters on [68Ga]Ga-DOTATOC PET/CT in neuroendocrine tumors (NET) patients treated with peptide receptor radionuclide therapy (PRRT). METHODS We retrospectively evaluated 39 NET patients (21 male, 18 female; mean age 60.7 y) within the FENET-2016 trial (CTiD:NCT04790708). PRRT was proposed with [177Lu]Lu-DOTATOC alone or combined with [90Y]Y-DOTATOC. [68Ga]Ga-DOTATOC PET/CT was performed at baseline and 3 months after PRRT. For each PET/CT, we calculated SUVmax, SUVmean, somatostatin receptor expressing tumor volume (SRETV), and total lesion somatostatin receptor expression (TLSRE), as well as their percentage of changes (Δ), both for liver (_L) and for total tumor burden (_WB). Early clinical response (3 months after PRRT) and PFS were evaluated according to RECIST 1.1 and institutional NET board. RESULTS Early clinical response identified 9 partial response (PR), 25 stable disease (SD), and 5 progressive disease (PD). Post-SRETV_WB and ΔSRETV_WB were progressively increased among response groups (p = 0.02 and p = 0.03, respectively). Likewise, median post-SRETV_L was significantly higher in PD patients (p = 0.03). SUVmax and TLSRE did not correlate with early clinical response. Median PFS was 31 months. Patients with ΔSRETV_WB lower than -4.17% as well as those with post-SRETV_WB lower than 34.8 cm3 showed a longer PFS (p = 0.006 and p = 0.06, respectively). Finally, multivariate analysis identified ΔSRETV_WB as an independent predictor for PFS. CONCLUSIONS our results could strengthen the importance of evaluating the burden of disease on [68Ga]Ga-DOTATOC PET/CT in NET patients treated with PRRT.
Collapse
|
5
|
Pineau J, Lima LMP, Platas‐Iglesias C, Zeevaart JR, Driver CHS, Le Bris N, Tripier R. Relevance of Palladium to Radiopharmaceutical Development Considering Enhanced Coordination Properties of TE1PA. Chemistry 2022; 28:e202200942. [PMID: 35560962 PMCID: PMC9401000 DOI: 10.1002/chem.202200942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Indexed: 11/09/2022]
Abstract
The limited use of palladium‐103 and ‐109 radionuclides for molecular radiotherapy is surely due to the lack of appropriate ligands capable of fulfilling all criteria required for application in nuclear medicine. Furthermore, the thermodynamic properties of these complexes in solution remain difficult to establish. The challenge is compounded when considering that radiolabeling of compounds for translation to clinical trials requires fast complexation. Thus, the coordination of Pd(II) and 103/109Pd‐nuclides is a huge challenge in terms of molecular design and physicochemical characterization. Herein, we report a comprehensive study highlighting TE1PA, a monopicolinate cyclam – already established in nuclear imaging with 64Cu‐PET (positron emission tomography) imaging tracers – as a highly relevant chelator for natural Pd and subsequently 109Pd‐nuclide. The structural, thermodynamic, kinetic and radiolabeling studies of Pd(II) with TE1PA, as well as the comparison of this complex with three structurally related derivatives, support palladium‐TE1PA radiopharmaceuticals as leading candidates for targeted nuclear medicine.
Collapse
Affiliation(s)
- Julie Pineau
- Univ Brest, UMR CNRS 6521 CEMCA 6 avenue Victor le Gorgeu 29238 Brest France) E-mail: s
| | - Luís M. P. Lima
- Instituto de Tecnologia Química e Biológica António Xavier Universidade Nova de Lisboa Av. da República 2780-157 Oeiras Portugal
| | - Carlos Platas‐Iglesias
- Departamento de Química Facultade de Ciencias & Centro de Investigaciones Científicas Avanzadas (CICA) Universidade da Coruña San Vicente de Elviña 15071 A Coruña Spain
| | - Jan Rijn Zeevaart
- South African Nuclear Energy Corporation Radiochemistry and PreClinical Imaging Facility Elias Motsoaledi Street, R104 Pelindaba North West 0240 South Africa
| | - Cathryn H. S. Driver
- South African Nuclear Energy Corporation Radiochemistry and PreClinical Imaging Facility Elias Motsoaledi Street, R104 Pelindaba North West 0240 South Africa
| | - Nathalie Le Bris
- Univ Brest, UMR CNRS 6521 CEMCA 6 avenue Victor le Gorgeu 29238 Brest France) E-mail: s
| | - Raphaël Tripier
- Univ Brest, UMR CNRS 6521 CEMCA 6 avenue Victor le Gorgeu 29238 Brest France) E-mail: s
| |
Collapse
|
6
|
de Aguiar Ferreira C, Heidari P, Ataeinia B, Sinevici N, Granito A, Kumar HM, Wehrenberg-Klee E, Mahmood U. Immune Checkpoint Inhibitor-Mediated Cancer Theranostics with Radiolabeled Anti-Granzyme B Peptide. Pharmaceutics 2022; 14:1460. [PMID: 35890355 PMCID: PMC9325142 DOI: 10.3390/pharmaceutics14071460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/25/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Although immune checkpoint inhibitors (ICI) have revolutionized cancer management, patient response can be heterogeneous, and the development of ICI resistance is increasingly reported. Novel treatment strategies are necessary not only to expand the use of ICI to previously unresponsive tumor types but also to overcome resistance. Targeted radionuclide therapy may synergize well with ICIs since it can promote a pro-inflammatory tumor microenvironment. We investigated the use of a granzyme B targeted peptide (GZP) as a cancer theranostic agent, radiolabeled with 68Ga (68Ga-GZP) as a PET imaging agent and radiolabeled with 90Y (90Y-GZP) as a targeted radionuclide therapy agent for combinational therapy with ICI in murine models of colon cancer. Our results demonstrate that GZP increasingly accumulates in tumor tissue after ICI and that the combination of ICI with 90Y-GZP promotes a dose-dependent response, achieving curative response in some settings and increased overall survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Umar Mahmood
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (C.d.A.F.); (P.H.); (B.A.); (N.S.); (A.G.); (H.M.K.); (E.W.-K.)
| |
Collapse
|
7
|
Amjed N, Naz A, Wajid A, Aslam M, Ahmad I. Nuclear model analysis and optimization of production data of the medically interesting 66,67,68 Ga via alpha induced reactions on 63,65 Cu targets. Appl Radiat Isot 2022; 188:110379. [DOI: 10.1016/j.apradiso.2022.110379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022]
|
8
|
Desai P, Rimal R, Sahnoun SEM, Mottaghy FM, Möller M, Morgenroth A, Singh S. Radiolabeled Nanocarriers as Theranostics-Advancement from Peptides to Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200673. [PMID: 35527333 DOI: 10.1002/smll.202200673] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/15/2022] [Indexed: 06/14/2023]
Abstract
Endogenous targeted radiotherapy is emerging as an integral modality to treat a variety of cancer entities. Nevertheless, despite the positive clinical outcome of the treatment using radiolabeled peptides, small molecules, antibodies, and nanobodies, a high degree of hepatotoxicity and nephrotoxicity still persist. This limits the amount of dose that can be injected. In an attempt to mitigate these side effects, the use of nanocarriers such as nanoparticles (NPs), dendrimers, micelles, liposomes, and nanogels (NGs) is currently being explored. Nanocarriers can prolong circulation time and tumor retention, maximize radiation dosage, and offer multifunctionality for different targeting strategies. In this review, the authors first provide a summary of radiation therapy and imaging and discuss the new radiotracers that are used preclinically and clinically. They then highlight and identify the advantages of radio-nanomedicine and its potential in overcoming the limitations of endogenous radiotherapy. Finally, the review points to the ongoing efforts to maximize the use of radio-nanomedicine for efficient clinical translation.
Collapse
Affiliation(s)
- Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Sabri E M Sahnoun
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
- Department of Radiology and Nuclear Medicine, School for Cardiovascular Diseases (CARIM) and School of oncology (GROW), Maastricht University, Maastricht, 6229 HX, The Netherlands
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Smriti Singh
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
- Max-Planck-Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| |
Collapse
|
9
|
Hartrampf P, Werner R, Buck A. Theranostics bei gut bis mäßig differenzierten GEP-NEN. Zentralbl Chir 2022; 147:249-255. [DOI: 10.1055/a-1826-3423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
ZusammenfassungNeuroendokrine Neoplasien (NEN) sind seltene, heterogene und typischerweise langsam wachsende Tumoren. Die häufigsten Lokalisationen finden sich im gastro-entero-pankreatischen System
(GEP-NEN). NENs werden nach proliferativer Aktivität (Ki-67-Index) eingeteilt (G1–3). Gut differenzierte Tumoren exprimieren dabei typischerweise Somatostatinrezeptoren (SSTR), die als
Zielstruktur in der nuklearmedizinischen Theranostik dienen. Bei diesem Prinzip kann nach einer diagnostischen molekularen Bildgebung, meist mittels
Positronenemissionstomografie/Computertomografie (PET/CT), eine individuell zugeschnittene Peptidradiorezeptortherapie (PRRT) mit einem β-Strahler-markierten Radiopharmakon erfolgen. In
Metaanalysen zeigte die Diagnostik mittels SSTR-gerichteter PET/CT eine Sensitivität von 93% und eine Spezifität von 96%. Die SSTR-gerichtete Diagnostik kann auch zur radioaktiven Markierung
von Tumoren verwendet werden, um eine zielgerichtete Chirurgie zu ermöglichen. Die Indikation zur Einleitung einer PRRT soll stets in einer interdisziplinären Tumorkonferenz getroffen
werden. Ein Tumorprogress unter der vorangegangenen Therapie sollte dokumentiert sein. Die Therapie wird intravenös und insgesamt 4-mal in 8-wöchigem Abstand in spezialisierten
nuklearmedizinischen Zentren verabreicht. Die Wirksamkeit der PRRT wurde in der NETTER-1-Studie prospektiv untersucht und konnte eine signifikante Verbesserung des progressionsfreien
Überlebens (primärer Endpunkt) zeigen. Ausgehend von diesen Studienergebnissen steht mit Lutathera (177Lu-DOTATATE) inzwischen ein in Deutschland zugelassenes Radiopharmazeutikum zu
Behandlung von nicht resektablen oder metastasierten bzw. progredienten, gut differenzierten (G1 und G2), SSTR-positiven GEP-NEN zur Verfügung.
Collapse
Affiliation(s)
- Philipp Hartrampf
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Rudolf Werner
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Andreas Buck
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Deutschland
| |
Collapse
|
10
|
Lepareur N. Cold Kit Labeling: The Future of 68Ga Radiopharmaceuticals? Front Med (Lausanne) 2022; 9:812050. [PMID: 35223907 PMCID: PMC8869247 DOI: 10.3389/fmed.2022.812050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last couple of decades, gallium-68 (68Ga) has gained a formidable interest for PET molecular imaging of various conditions, from cancer to infection, through cardiac pathologies or neuropathies. It has gained routine use, with successful radiopharmaceuticals such as somatostatin analogs ([68Ga]Ga-DOTATOC and [68Ga]GaDOTATATE) for neuroendocrine tumors, and PSMA ligands for prostate cancer. It represents a major clinical impact, particularly in the context of theranostics, coupled with their 177Lu-labeled counterparts. Beside those, a bunch of new 68Ga-labeled molecules are in the preclinical and clinical pipelines, with some of them showing great promise for patient care. Increasing clinical demand and regulatory issues have led to the development of automated procedures for the production of 68Ga radiopharmaceuticals. However, the widespread use of these radiopharmaceuticals may rely on simple and efficient radiolabeling methods, undemanding in terms of equipment and infrastructure. To make them technically and economically accessible to the medical community and its patients, it appears mandatory to develop a procedure similar to the well-established kit-based 99mTc chemistry. Already available commercial kits for the production of 68Ga radiopharmaceuticals have demonstrated the feasibility of using such an approach, thus paving the way for more kit-based 68Ga radiopharmaceuticals to be developed. This article discusses the development of 68Ga cold kit radiopharmacy, including technical issues, and regulatory aspects.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer), UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
11
|
Naik M, Al-Nahhas A, Khan SR. Treatment of Neuroendocrine Neoplasms with Radiolabeled Peptides-Where Are We Now. Cancers (Basel) 2022; 14:761. [PMID: 35159027 PMCID: PMC8833798 DOI: 10.3390/cancers14030761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been one of the most successful and exciting examples of theranostics in nuclear medicine in recent decades and is now firmly embedded in many treatment algorithms for unresectable or metastatic neuroendocrine neoplasms (NENs) worldwide. It is widely considered to be an effective treatment for well- or moderately differentiated neoplasms, which express high levels of somatostatin receptors that can be selectively targeted. This review article outlines the scientific basis of PRRT in treatment of NENs and describes its discovery dating back to the early 1990s. Early treatments utilizing Indium-111, a γ-emitter, showed promise in reduction in tumor size and improvement in biochemistry, but were also met with high radiation doses and myelotoxic and nephrotoxic effects. Subsequently, stable conjugation of DOTA-peptides with β-emitting radionuclides, such as Yttrium-90 and Lutetium-177, served as a breakthrough for PRRT and studies highlighted their potential in eliciting progression-free survival and quality of life benefits. This article will also elaborate on the key trials which paved the way for its approval and will discuss therapeutic considerations, such as patient selection and administration technique, to optimize its use.
Collapse
Affiliation(s)
- Mitesh Naik
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| | | | - Sairah R. Khan
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| |
Collapse
|
12
|
Pijarowska-Kruszyna J, Pocięgiel M, Mikołajczak R. Radionuclide generators. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
13
|
Youssef A, Haskali MB, Gorringe KL. The Protein Landscape of Mucinous Ovarian Cancer: Towards a Theranostic. Cancers (Basel) 2021; 13:5596. [PMID: 34830751 PMCID: PMC8616050 DOI: 10.3390/cancers13225596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/20/2023] Open
Abstract
MOC is a rare histotype of epithelial ovarian cancer, and current management options are inadequate for the treatment of late stage or recurrent disease. A shift towards personalised medicines in ovarian cancer is being observed, with trials targeting specific molecular pathways, however, MOC lags due to its rarity. Theranostics is a rapidly evolving category of personalised medicine, encompassing both a diagnostic and therapeutic approach by recognising targets that are expressed highly in tumour tissue in order to deliver a therapeutic payload. The present review evaluates the protein landscape of MOC in recent immunohistochemical- and proteomic-based research, aiming to identify potential candidates for theranostic application. Fourteen proteins were selected based on cell membrane localisation: HER2, EGFR, FOLR1, RAC1, GPR158, CEACAM6, MUC16, PD-L1, NHE1, CEACAM5, MUC1, ACE2, GP2, and PTPRH. Optimal proteins to target using theranostic agents must exhibit high membrane expression on cancerous tissue with low expression on healthy tissue to afford improved disease outcomes with minimal off-target effects and toxicities. We provide guidelines to consider in the selection of a theranostic target for MOC and suggest future directions in evaluating the results of this review.
Collapse
Affiliation(s)
- Arkan Youssef
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Mohammad B. Haskali
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Kylie L. Gorringe
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia;
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
14
|
Uccelli L, Boschi A, Cittanti C, Martini P, Panareo S, Tonini E, Nieri A, Urso L, Caracciolo M, Lodi L, Carnevale A, Giganti M, Bartolomei M. 90Y/ 177Lu-DOTATOC: From Preclinical Studies to Application in Humans. Pharmaceutics 2021; 13:1463. [PMID: 34575538 PMCID: PMC8469896 DOI: 10.3390/pharmaceutics13091463] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
The PRRT (Peptide Receptor Radionuclide Therapy) is a promising modality treatment for patients with inoperable or metastatic neuroendocrine tumors (NETs). Progression-free survival (PFS) and overall survival (OS) of these patients are favorably comparable with standard therapies. The protagonist in this type of therapy is a somatostatin-modified peptide fragment ([Tyr3] octreotide), equipped with a specific chelating system (DOTA) capable of creating a stable bond with β-emitting radionuclides, such as yttrium-90 and lutetium-177. In this review, covering twenty five years of literature, we describe the characteristics and performances of the two most used therapeutic radiopharmaceuticals for the NETs radio-treatment: [90Y]Y-DOTATOC and [177Lu]Lu-DOTATOC taking this opportunity to retrace the most significant results that have determined their success, promoting them from preclinical studies to application in humans.
Collapse
Affiliation(s)
- Licia Uccelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Petra Martini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
| | - Stefano Panareo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Eugenia Tonini
- Medical Physics Unit, University Hospital, 44124 Ferrara, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Urso
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Matteo Caracciolo
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Luca Lodi
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Melchiore Giganti
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.U.); (A.C.); (M.G.)
- Radiology Unit, University Hospital, 44124 Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, University Hospital, 44124 Ferrara, Italy; (S.P.); (A.N.); (L.U.); (M.C.); (L.L.); (M.B.)
| |
Collapse
|
15
|
Rennert J, Grosse J, Einspieler I, Bäumler W, Stroszczynski C, Jung EM. Complementary imaging of ultrasound and PET/CT: A new opportunity? Clin Hemorheol Microcirc 2021; 79:39-54. [PMID: 34420945 DOI: 10.3233/ch-219105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM To evaluate the effectiveness of complementary imaging of high-resolution ultrasound including CEUS with PET/CT for tissue characterization and tumor detection. MATERIAL AND METHODS 100 patients were examined with PET/CT and US/CEUS between January 2018 until February 2020. All patients underwent PET/CT followed by selective US/CEUS within 4 weeks. Comparison regarding concordant or diverging findings in PET/CT and US. Analysis of the differences concerning the lesions number of found by PET/CT and US/CEUS or the possibility of a secured diagnosis following ultrasound causing therapeutic changes. RESULTS Diverging findings regarding the number of liver lesions in PET/CT and CEUS were found in 35 out of 64 patients (54%). Regarding renal lesions, a more definite diagnosis following ultrasound, causing a change of therapeutic approach, was achieved in 89%. Concordant results in PET/CT and US were found in 83% of patients with splenic and nodal findings. In 78% of patients with increased musculoskeletal or soft tissue tracer uptake, US was able to make a secured diagnosis with therapeutic changes. CONCLUSION The present results indicate a strong benefit of complementary imaging of PET/CT and selective, high-resolution ultrasound especially in patients with liver, renal and musculoskeletal or soft tissue findings.
Collapse
Affiliation(s)
- Janine Rennert
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Jirka Grosse
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ingo Einspieler
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Wolf Bäumler
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | | | - Ernst Michael Jung
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
16
|
Satpati D. Recent Breakthrough in 68Ga-Radiopharmaceuticals Cold Kits for Convenient PET Radiopharmacy. Bioconjug Chem 2021; 32:430-447. [PMID: 33630583 DOI: 10.1021/acs.bioconjchem.1c00010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
68Ga-PET has emerged as an important diagnostic tool for precise detection and monitoring of oncological situations. Availability, cost, and radiosynthesis procedure are determining steps for success of a radioisotope/radiopharmaceutical in nuclear medicine. Availability of 68Ga from a 68Ge/68Ga generator containing a long-lived parent radioisotope (68Ge: t1/2 = 271 days) and an inexpensive, simplified production of 68Ga-radiopharmaceuticals through kit methodology has allowed smooth accommodation of 68Ga-PET in clinics. The uncomplicated formulation of 68Ga-radiopharmaceuticals from a lyophilized, cold kit is an impending breakthrough in clinical PET. The huge success of 68Ga in neuroendocrine tumor and prostate cancer imaging along with the regulatory approval of respective cold kits has opened a pathway for development of kits for other evolving radiotracers. There is a definite scope for increased participation of commercial manufacturers and distributors of cold kits to spread the potential of 68Ga worldwide across all the geographical locations and satellite centers.
Collapse
Affiliation(s)
- Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai-400085, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| |
Collapse
|
17
|
Aslam MT, Ali W, Hussain M. Nuclear model analysis of the 65Cu(α, n) 68Ga reaction for the production of 68Ga up to 40 MeV. Appl Radiat Isot 2021; 170:109590. [PMID: 33493791 DOI: 10.1016/j.apradiso.2021.109590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 01/27/2023]
Abstract
The charge particle (α) induced reactions on enriched copper (65Cu) are investigated for the production of 68Ga. The data sets of experimental cross sections are compiled, normalized and nuclear model analysis is done using calculational codes namely, ALICE-IPPE, TALYS 1.95 and EMPIRE 3.2. The theoretical production cross sections via alpha particle induced reactions are calculated to present a set of recommended cross sections. The calculated cross sections are utilized to deduce thick target yield (TTY) for the 65Cu (α, n) 68Ga reaction. The range of energy for production of 68Ga is suggested up to 40 MeV having least contribution of radio-impurities.
Collapse
Affiliation(s)
| | - Waris Ali
- Department of Physics, Government Islamia College, Civil Lines, Lahore, 54000, Lahore, Pakistan
| | - Mazhar Hussain
- Department of Physics, Government College University, Lahore, 54000, Lahore, Pakistan
| |
Collapse
|
18
|
Hall AJ, Haskali MB. Radiolabelled Peptides: Optimal Candidates for Theranostic Application in Oncology. Aust J Chem 2021. [DOI: 10.1071/ch21118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Methods to radiolabel somatostatin analogs with [18F]fluoride: current status, challenges, and progress in clinical applications. J Radioanal Nucl Chem 2020. [DOI: 10.1007/s10967-020-07437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
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:E4012. [PMID: 32887456 PMCID: PMC7504749 DOI: 10.3390/molecules25174012] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [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
|
21
|
Chauhan A, El-Khouli R, Waits T, Agrawal R, Siddiqui F, Tarter Z, Horn M, Weiss H, Oates E, Evers BM, Anthony L. Post FDA approval analysis of 200 gallium-68 DOTATATE imaging: A retrospective analysis in neuroendocrine tumor patients. Oncotarget 2020; 11:3061-3068. [PMID: 32850010 PMCID: PMC7429177 DOI: 10.18632/oncotarget.27695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
Gallium-68 DOTATATE provides physiologic imaging and assists in disease localization for somatostatin receptor (SSTR) positive neuroendocrine tumor (NET) patients. However, questions regarding usefulness of gallium- 68 DOTATATE imaging in identifying the primary site in neuroendocrine tumors (NETS) of unknown primary, correlation of NET grade with median Standardized Uptake Value (SUV) and effects of long acting somatostatin analog on gallium-68 DOTATATE imaging quality needs to be evaluated. A single institution retrospective review of the first 200 NET patients with gallium-68 DOTATATE imaging from Dec 2016 to Dec 2017 was conducted. Questions related to NETs of unknown primary, correlation of Standardized Uptake Value (SUV) to Ki-67 (which signifies proliferation rate), the effects of long-acting systemic somatostatin analog (SSA) on SUV were part of our data analysis. From these 200 patients, 59.5% (119) were females, 40.5% (81) were males; the median age was 62 years. The following primary tumor sites were identified: small bowel-37.5%; pancreas-18.5%; bronchial-14%; colon-3.5%; rectum-2%; appendix-1.5%; adrenal-0.5%; prostate-0.5%; others-3% and unknown primary-19%. Mean hepatic SUV of the lesion with the greatest radiolabeled uptake in 96 patients was similar irrespective to exposure to long acting SSA. Patients exposed to long acting SSA had mean SUV of 31.3 vs 27.8 for SSA naïve patients. The difference was not statistically significant. Gallium-68 DOTATATE imaging seems to distinguished G3 NET from G1/G2 based on mean SUV, and also identified the primary tumor site in 17 of 38 (45%) patients with unknown primary. Systemic exposure to long acting SSA does not appear to influence mean SUV of gallium-68 DOTATATE scan.
Collapse
Affiliation(s)
- Aman Chauhan
- Department of Internal Medicine-Medical Oncology and the Markey Cancer Center, University of Kentucky, Lexington, KY, USA
- Co primary authors
| | - Riham El-Khouli
- Department of Radiology, University of Kentucky, Lexington, KY, USA
- Co primary authors
| | - Timothy Waits
- Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - Rohitashva Agrawal
- Department of Internal Medicine-Medical Oncology and the Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Fariha Siddiqui
- College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Zachary Tarter
- College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Millicent Horn
- College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Heidi Weiss
- Department of Biostatistics and the Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Oates
- Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - B. Mark Evers
- Department of Surgery and the Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Lowell Anthony
- Department of Internal Medicine-Medical Oncology and the Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
22
|
Werner RA, Hänscheid H, Leal JP, Javadi MS, Higuchi T, Lodge MA, Buck AK, Pomper MG, Lapa C, Rowe SP. Impact of Tumor Burden on Quantitative [ 68Ga] DOTATOC Biodistribution. Mol Imaging Biol 2020; 21:790-798. [PMID: 30406512 DOI: 10.1007/s11307-018-1293-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE As has been previously reported, the somatostatin receptor (SSTR) imaging agent [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotate ([68Ga]DOTATATE) demonstrates lower uptake in normal organs in patients with a high neuroendocrine tumor (NET) burden. Given the higher SSTR affinity of [68Ga] DOTATATE, we aimed to quantitatively investigate the biodistribution of [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotide ([68Ga]DOTATOC) to determine a potential correlation between uptake in normal organs and NET burden. PROCEDURES Of the 44 included patients, 36/44 (82 %) patients demonstrated suspicious radiotracer uptake on [68Ga] DOTATOC positron emission tomography (PET)/X-ray computed tomography (CT). Volumes of interest (VOIs) were defined for tumor lesions and normal organs (spleen, liver, kidneys, adrenals). Mean body weight corrected standardized uptake value (SUVmean) for normal organs was assessed and was used to calculate the corresponding mean specific activity uptake (Upt: fraction of injected activity per kg of tissue). For the entire tumor burden, SUVmean, maximum standardized uptake value (SUVmax), and the total mass (TBM) was calculated and the decay corrected tumor fractional uptake (TBU) was assessed. A Spearman's rank correlation coefficient was used to determine the correlations between normal organ uptake and tumor burden. RESULTS The median SUVmean was 18.7 for the spleen (kidneys, 9.2; adrenals, 6.8; liver, 5.6). For tumor burden, the median values were SUVmean 6.9, SUVmax 35.5, TBM 42.6 g, and TBU 1.2 %. With increasing volume of distribution, represented by lean body mass and body surface area (BSA), Upt decreased in kidneys, liver, and adrenal glands and SUVmean increased in the spleen. Correlation improved only for both kidneys and adrenals when the influence of the tumor uptake on the activity available for organ uptake was taken into account by the factor 1/(1-TBU). TBU was neither predictive for SUVmean nor for Upt in any of the organs. The distribution of organ Upt vs. BSA/(1-TBU) were not different for patients with minor TBU (<3 %) vs. higher TBU (>7 %), indicating that the correlations observed in the present study are explainable by the body size effect. High tumor mass and uptake mitigated against G1 NET. CONCLUSIONS There is no significant impact on normal organ biodistribution with increasing tumor burden on [68Ga] DOTATOC PET/CT. Potential implications include increased normal organ dose with [177Lu-DOTA]0-D-Phe1-Tyr3-Octreotide and decreased absolute lesion detection with [68Ga] DOTATOC in high NET burden.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany.,The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Heribert Hänscheid
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jeffrey P Leal
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Mehrbod S Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin A Lodge
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA. .,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
The efficacy of 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) in patients with metastatic neuroendocrine tumours: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2020; 146:1533-1543. [PMID: 32281025 DOI: 10.1007/s00432-020-03181-2] [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] [Received: 09/17/2019] [Accepted: 03/11/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE To evaluate the efficacy of 177Lu-DOTA0-Tyr3-octreotate (177Lu-DOTATATE) radionuclide therapy in patients with inoperable or metastatic neuroendocrine tumours (NETs), (PROSPERO ID CRD42019130755). METHODS All published clinical studies of NETs treated with 177Lu-DOTATATE were identified based on systematic searches in the PubMed, EMBASE, Cochrane Library, Web of Science and ClinicalTrials.gov databases up to January 2019. Among these studies, only the reports evaluated with the "Response Evaluation Criteria in Solid Tumours (RECIST)" or "Southwest Oncology Group (SWOG)" criteria or both were included. We analysed the disease response rate (DRR) and disease control rate (DCR) of each group to evaluate the efficacy of 177Lu-DOTATATE. RESULTS Fifteen studies were selected from 715 references. The pooled effect in the RECIST group (13 studies) was 27.58% (95% confidence interval (CI) 21.03-35.27%) for the DRR and 79.14% (95% CI 75.83-82.1%) for the DCR. In the SWOG criteria group (7 studies), the pooled effect was 20.59% (95% CI 10.89-35.51%) for the DRR and 78.28% (95% CI 74.39-81.72%) for the DCR. Therefore, the RECIST and SWOG groups showed similar DRRs and DCRs after177Lu-DOTATATE treatment, indicating that 177Lu-DOTATATE treatment has excellent efficacy with a control rate of approximately 78-79%. Moreover, adverse effects of 177Lu-DOTATATE were minimal, including fatigue, nausea, vomiting and hormonal disorders. CONCLUSIONS For patients with inoperable or metastatic NETs, 177Lu-DOTATATE is an effective treatment with minimal side effects.
Collapse
|
24
|
Cox CPW, Segbers M, Graven LH, Brabander T, van Assema DME. Standardized image quality for 68Ga-DOTA-TATE PET/CT. EJNMMI Res 2020; 10:27. [PMID: 32201912 PMCID: PMC7085989 DOI: 10.1186/s13550-020-0601-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/31/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Positron emission tomography (PET) imaging with 68Gallium labeled somatostatin analogues (68Ga-DOTA-SSA) plays a key role in neuroendocrine tumor management. The impact of patient size on PET image quality is not well known for PET imaging with 68Ga-DOTA-SSA. The aim of this study is to propose a dose regimen based on patient size that optimizes image quality and yields sufficient image quality for diagnosis. METHODS Twenty-one patients (12 males, 9 females) were prospectively included for 68Gallium-DOTA-Tyr3-Octreotate (68Ga-DOTA-TATE) PET/CT, which was acquired in whole body list mode using 6 min per bed position (mbp). The list-mode events were randomly sampled to obtain 1 to 6 mbp PET reconstructions. For semi-quantitative assessment of image quality, the signal-to-noise ratio (SNR) was measured in the liver. The SNR normalized (SNRnorm) for administered activity and mbp was correlated with body mass, length, body mass index, body mass/length, and lean body mass. Three experienced nuclear medicine physicians visually graded image quality using a 4-point scale, and categorically scored the number of somatostatin-receptor positive lesions for each reconstruction. To investigate the impact of image quality on lesion quantification, the mean, maximum, and peak standardized uptake values (SUVs) of one abdominal lesion were measured in the 1 to 6 mbp PET reconstructions. RESULTS Of all patient-dependent parameters, body mass showed the strongest correlation (R2 = 0.6) with SNRnorm. Lesion detectability analysis showed no significant difference for 3-5 mbp compared with the complete 6 mbp PET reconstruction. The SUV measurements showed no significant (p > 0.05) differences across the reconstructions. Visual assessment revealed that an SNR of 6.2 results in PET scans with moderate to good image quality. A non-linear expression was derived to calculate the required (dose × acquisition time) product (DTP) for the chosen SNR level of 6.2 that would yield a more constant image quality. CONCLUSION Body mass can be used to predict 68Ga-DOTA-TATE PET image quality. The proposed non-linear dose regimen based on body mass standardizes the image quality while maintaining sufficient image quality for diagnosis.
Collapse
Affiliation(s)
- Christina P. W. Cox
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, The Netherlands
| | - Marcel Segbers
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, The Netherlands
| | - Laura H. Graven
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, The Netherlands
| | - Tessa Brabander
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, The Netherlands
| | - Daniëlle M. E. van Assema
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
25
|
Rousseau E, Lau J, Zhang Z, Zhang C, Kwon D, Uribe CF, Kuo HT, Zeisler J, Bratanovic I, Lin KS, Bénard F. Comparison of biological properties of [ 177 Lu]Lu-ProBOMB1 and [ 177 Lu]Lu-NeoBOMB1 for GRPR targeting. J Labelled Comp Radiopharm 2020; 63:56-64. [PMID: 31715025 DOI: 10.1002/jlcr.3815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 11/06/2022]
Abstract
The gastrin-releasing peptide receptor (GRPR) is overexpressed in prostate cancer and other solid malignancies. Following up on our work on [68 Ga]Ga-ProBOMB1 that had better imaging characteristics than [68 Ga]Ga-NeoBOMB1, we investigated the effects of substituting 68 Ga for 177 Lu to determine if the resulting radiopharmaceuticals could be used with a therapeutic aim. We radiolabeled the bombesin antagonist ProBOMB1 (DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-ψ-Pro-NH2 ) with lutetium-177 and compared it with [177 Lu]Lu-NeoBOMB1 (obtained in 54.2 ± 16.5% isolated radiochemical yield with >96% radiochemical purity and 440.8 ± 165.1 GBq/μmol molar activity) for GRPR targeting. Lu-NeoBOMB1 had better binding affinity for GRPR than Lu-ProBOMB1 (Ki values: 2.26 ± 0.24 and 30.2 ± 3.23nM). [177 Lu]Lu-ProBOMB1 was obtained in 53.7 ± 5.4% decay-corrected radiochemical yield with 444.2 ± 193.2 GBq/μmol molar activity and >95% radiochemical purity. In PC-3 prostate cancer xenograft mice, tumor uptake of [177 Lu]Lu-ProBOMB1 was 3.38 ± 1.00, 1.32 ± 0.24, and 0.31 ± 0.04%ID/g at 1, 4, and 24 hours pi. However, the uptake in tumor was lower than [177 Lu]Lu-NeoBOMB1 at all time points. [177 Lu]Lu-ProBOMB1 was inferior to [177 Lu]Lu-NeoBOMB1, which had better therapeutic index for the organs receiving the highest doses.
Collapse
Affiliation(s)
- Etienne Rousseau
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Joseph Lau
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Zhengxing Zhang
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Chengcheng Zhang
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Daniel Kwon
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Carlos F Uribe
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Hsiou-Ting Kuo
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Jutta Zeisler
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Ivica Bratanovic
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Werner RA, Chen X, Rowe SP, Lapa C, Javadi MS, Higuchi T. Recent paradigm shifts in molecular cardiac imaging—Establishing precision cardiology through novel 18F-labeled PET radiotracers. Trends Cardiovasc Med 2020; 30:11-19. [DOI: 10.1016/j.tcm.2019.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/30/2022]
|
27
|
Mirzaei S, Revheim ME, Raynor W, Zehetner W, Knoll P, Zandieh S, Alavi A. 64Cu-DOTATOC PET-CT in Patients with Neuroendocrine Tumors. Oncol Ther 2019; 8:125-131. [PMID: 32700066 PMCID: PMC7360020 DOI: 10.1007/s40487-019-00104-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction Several radiolabeled somatostatin analogues have been developed for molecular imaging of neuroendocrine tumors (NETs) with single-photon emission computed tomography (SPECT) and positron-emission tomography (PET). The aim of the present study was to report our first results using 64Cu-DOTATOC in patients with NETs. Methods Thirty-three patients with NETs (15 female, 18 male; mean age 64 ± 13 years) were included in this retrospective study. 64Cu-DOTATOC PET–CT scans were performed on all patients. Results Five out of 33 patients with a history of NET after surgical removal of the primary lesion showed no pathological lesions on PET–CT imaging and 8/33 patients had enhanced uptake in the area of recurrent meningioma at the skull base. The remaining 20/33 patients had a history of neuroendocrine tumor in the gastrointestinal tract (GEP-NET) and were presented with at least one pathological lesion. Conclusion The high detection rate of suspected lesions in patients with NETs and the high target-to-background contrast found in this study hold promise for the safe application of 64Cu-DOTATOC in patients with NET.
Collapse
Affiliation(s)
- Siroos Mirzaei
- Department of Nuclear Medicine with PET-Center, Wilhelminenspital, Vienna, Austria.
| | - Mona-Eilsabeth Revheim
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - William Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Walter Zehetner
- Department of Nuclear Medicine with PET-Center, Wilhelminenspital, Vienna, Austria
| | - Peter Knoll
- Department of Nuclear Medicine with PET-Center, Wilhelminenspital, Vienna, Austria
| | - Shahin Zandieh
- Department of Radiology and Nuclear Medicine, Hanusch Hospital, Vienna, Austria.,Department of Radiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
28
|
Preparation of [ 177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 for radiotherapy of insulinoma: a detailed insight into the radiochemical intricacies. Nucl Med Biol 2019; 78-79:31-40. [PMID: 31731177 DOI: 10.1016/j.nucmedbio.2019.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/11/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION [177Lu]Lu-DOTA-Ahx-Lys40-Exendin-4 ([177Lu]Lu-DOTA-Exendin-4) is a potential agent for radiotherapy of insulinomas owing to its specificity towards GLP-1 (Glucagon like peptide-1) receptors over-expressed on such cancers. The objective of the present study is to optimize the various radiochemistry parameters for the consistent formulation of the agent with high radiolabeling yield using carrier added [177Lu]LuCl3 and also to evaluate its biological behaviour in small animal model. METHODS In order to optimize the radiolabeling parameters, DOTA-Exendin-4 was radiolabeled with [177Lu]LuCl3 in two different buffer systems (sodium acetate and HEPES) at three different temperatures (45, 65 and 95 °C) using three different ligand to metal ratios (3:1, 4:1 and 5:1). The radiolabeled peptide was characterized by both paper chromatography and HPLC. The effect of addition of three different radio-protectors on complexation yield was also studied. Bio-distribution studies were carried out in healthy Swiss mice to evaluate the pharmacokinetic behaviour of the radiolabeled peptide as well as to determine the in vivo specificity of the radiotracer towards GLP-1 receptors (blocking studies). Urine and kidney lysate of the animals were analyzed at various post-administration time-points in order to determine the in vivo stability of the radiolabeled peptide. RESULTS The [177Lu]Lu-DOTA-Exendin-4 complex could be prepared consistently with >95% radiolabeling yield using the optimized reaction conditions. Bio-distribution studies revealed early accumulation of [177Lu]Lu-DOTA-Exendin-4 in pancreas along with fast clearance via renal pathway. Significantly high accumulation of the radiotracer was observed in kidneys. Analyses of urine and kidney lysate of the animals revealed in vivo stability of [177Lu]Lu-DOTA-Exendin-4. Blocking studies showed displacement of significant amount of radiotracer from GLP-1 receptor-positive organs such as, pancreas and lungs (p <0.05) in presence of unlabeled peptide, indicating the specificity of the radiolabeled preparation towards GLP-1 receptors. CONCLUSIONS Present study shows that [177Lu]Lu-DOTA-Exendin-4 could be formulated for radiotherapeutic application with high radiochemical purity and adequate in vivo stability using [177Lu]LuCl3 produced via direct neutron irradiation. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Findings of the present study will be helpful in preparing the patient dose of [177Lu]Lu-labeled Exendin for radiotherapy of insulinoma using carrier added [177Lu]LuCl3, produced in a medium flux reactor, without the requirement of post-labeling purification.
Collapse
|
29
|
Schobert I, Chapiro J, Pucar D, Saperstein L, Savic LJ. Fluorodeoxyglucose PET for Monitoring Response to Embolotherapy (Transarterial Chemoembolization) in Primary and Metastatic Liver Tumors. PET Clin 2019; 14:437-445. [DOI: 10.1016/j.cpet.2019.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Werner RA, Thackeray JT, Pomper MG, Bengel FM, Gorin MA, Derlin T, Rowe SP. Recent Updates on Molecular Imaging Reporting and Data Systems (MI-RADS) for Theranostic Radiotracers-Navigating Pitfalls of SSTR- and PSMA-Targeted PET/CT. J Clin Med 2019; 8:E1060. [PMID: 31331016 PMCID: PMC6678732 DOI: 10.3390/jcm8071060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The theranostic concept represents a paradigmatic example of personalized treatment. It is based on the use of radiolabeled compounds which can be applied for both diagnostic molecular imaging and subsequent treatment, using different radionuclides for labelling. Clinically relevant examples include somatostatin receptor (SSTR)-targeted imaging and therapy for the treatment of neuroendocrine tumors (NET), as well as prostate-specific membrane antigen (PSMA)-targeted imaging and therapy for the treatment of prostate cancer (PC). As such, both classes of radiotracers can be used to triage patients for theranostic endoradiotherapy using positron emission tomography (PET). While interpreting PSMA- or SSTR-targeted PET/computed tomography scans, the reader has to navigate certain pitfalls, including (I.) varying normal biodistribution between different PSMA- and SSTR-targeting PET radiotracers, (II.) varying radiotracer uptake in numerous kinds of both benign and malignant lesions, and (III.) resulting false-positive and false-negative findings. Thus, two novel reporting and data system (RADS) classifications for PSMA- and SSTR-targeted PET imaging (PSMA- and SSTR-RADS) have been recently introduced under the umbrella term molecular imaging reporting and data systems (MI-RADS). Notably, PSMA- and SSTR-RADS are structured in a reciprocal fashion, i.e., if the reader is familiar with one system, the other system can readily be applied. Learning objectives of the present case-based review are as follows: (I.) the theranostic concept for the treatment of NET and PC will be briefly introduced, (II.) the most common pitfalls on PSMA- and SSTR-targeted PET/CT will be identified, (III.) the novel framework system for theranostic radiotracers (MI-RADS) will be explained, applied to complex clinical cases and recent studies in the field will be highlighted. Finally, current treatment strategies based on MI-RADS will be proposed, which will demonstrate how such a generalizable framework system truly paves the way for clinically meaningful molecular imaging-guided treatment of either PC or NET. Thus, beyond an introduction of MI-RADS, the present review aims to provide an update of recently published studies which have further validated the concept of structured reporting systems in the field of theranostics.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Martin G Pomper
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Michael A Gorin
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Steven P Rowe
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA.
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
31
|
Rozenblum L, Mokrane FZ, Yeh R, Sinigaglia M, Besson F, Seban RD, Chougnet CN, Revel-Mouroz P, Zhao B, Otal P, Schwartz LH, Dercle L. The role of multimodal imaging in guiding resectability and cytoreduction in pancreatic neuroendocrine tumors: focus on PET and MRI. Abdom Radiol (NY) 2019; 44:2474-2493. [PMID: 30980115 DOI: 10.1007/s00261-019-01994-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pancreatic neuroendocrine tumors (pNETs) are rare neoplasms that secrete peptides and neuro-amines. pNETs can be sporadic or hereditary, syndromic or non-syndromic with different clinical presentations and prognoses. The role of medical imaging includes locating the tumor, assessing its extent, and evaluating the feasibility of curative surgery or cytoreduction. Pancreatic NETs have very distinctive phenotypes on CT, MRI, and PET. PET have been demonstrated to be very sensitive to detect either well-differentiated pNETs using 68Gallium somatostatin receptor (SSTR) radiotracers, or more aggressive undifferentiated pNETS using 18F-FDG. A comprehensive interpretation of multimodal imaging guides resectability and cytoreduction in pNETs. The imaging phenotype provides information on the differentiation and proliferation of pNETs, as well as the spatial and temporal heterogeneity of tumors with prognostic and therapeutic implications. This review provides a structured approach for standardized reading and reporting of medical imaging studies with a focus on PET and MR techniques. It explains which imaging approach should be used for different subtypes of pNET and what a radiologist should be looking for and reporting when interpreting these studies.
Collapse
Affiliation(s)
- Laura Rozenblum
- Sorbonne Université, Service de Médecine Nucléaire, AP-HP, Hôpital La Pitié-Salpêtrière, 75013, Paris, France
| | - Fatima-Zohra Mokrane
- Radiology Department, Toulouse University Hospital, 1 Avenue du Professeur Jean Poulhes, 31059, Toulouse, France
- Department of Radiology, New York Presbyterian Hospital, Columbia University, New York, NY, USA
| | - Randy Yeh
- Memorial Sloan Kettering Cancer Center, Molecular Imaging and Therapy Service, New York, NY, USA
| | - Mathieu Sinigaglia
- Department of Imaging and Nuclear Medicine, Institut Claudius Regaud - Institut Universitaire du Cancer de Toulouse - Oncopole, Toulouse, France
| | - Florent Besson
- Paris Sud University, Kremlin Bicêtre Hospital, Paris, France
| | - Romain-David Seban
- Department of Nuclear Medicine, Institut Curie-René Huguenin, Saint-Cloud, France
| | - Cecile N Chougnet
- Department of Endocrine Oncology, Hôpital Saint Louis, Paris, France
| | - Paul Revel-Mouroz
- Radiology Department, Toulouse University Hospital, 1 Avenue du Professeur Jean Poulhes, 31059, Toulouse, France
| | - Binsheng Zhao
- Department of Radiology, New York Presbyterian Hospital, Columbia University, New York, NY, USA
| | - Philippe Otal
- Radiology Department, Toulouse University Hospital, 1 Avenue du Professeur Jean Poulhes, 31059, Toulouse, France
| | - Lawrence H Schwartz
- Department of Radiology, New York Presbyterian Hospital, Columbia University, New York, NY, USA
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University, New York, NY, USA.
- UMR 1015, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, 94805, France.
| |
Collapse
|
32
|
Rosecker V, Denk C, Maurer M, Wilkovitsch M, Mairinger S, Wanek T, Mikula H. Cross-Isotopic Bioorthogonal Tools as Molecular Twins for Radiotheranostic Applications. Chembiochem 2019; 20:1530-1535. [PMID: 30742739 PMCID: PMC6617999 DOI: 10.1002/cbic.201900042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Indexed: 11/08/2022]
Abstract
Radiotheranostics are designed by labeling targeting (bio)molecules with radionuclides for diagnostic or therapeutic application. Because the pharmacokinetics of therapeutic compounds play a pivotal role, chemically closely related imaging agents are used to evaluate the overall feasibility of the therapeutic approach. "Theranostic relatives" that utilize different elements are frequently used in clinical practice. However, variations in pharmacokinetics, biodistribution, and target affinity due to different chemical properties of the radioisotopes remain as hurdles to the design of optimized clinical tools. Herein, the design and synthesis of structurally identical compounds, either for diagnostic (18 F and a stable metal isotope) or therapeutic application (radiometal and stable 19 F), are reported. Such "molecular twins" have been prepared by applying a modular strategy based on click chemistry that enables efficient radiolabeling of compounds containing a metal complex and a tetrazine moiety. This additional bioorthogonal functionality can be used for subsequent radiolabeling of (bio)molecules or pretargeting approaches, which is demonstrated in vitro.
Collapse
Affiliation(s)
- Veronika Rosecker
- Institute of Applied Synthetic ChemistryTU Wien (Vienna University of Technology)Getreidemarkt 91060ViennaAustria
| | - Christoph Denk
- Institute of Applied Synthetic ChemistryTU Wien (Vienna University of Technology)Getreidemarkt 91060ViennaAustria
| | - Melanie Maurer
- Institute of Applied Synthetic ChemistryTU Wien (Vienna University of Technology)Getreidemarkt 91060ViennaAustria
| | - Martin Wilkovitsch
- Institute of Applied Synthetic ChemistryTU Wien (Vienna University of Technology)Getreidemarkt 91060ViennaAustria
| | - Severin Mairinger
- Preclinical Molecular ImagingAIT Austrian Institute of Technology2444SeibersdorfAustria
| | - Thomas Wanek
- Preclinical Molecular ImagingAIT Austrian Institute of Technology2444SeibersdorfAustria
| | - Hannes Mikula
- Institute of Applied Synthetic ChemistryTU Wien (Vienna University of Technology)Getreidemarkt 91060ViennaAustria
| |
Collapse
|
33
|
Vyas CK, Lee JY, Hur MG, Yang SD, Kong YB, Lee EJ, Park JH. Chitosan-TiO 2 composite: A potential 68Ge/ 68Ga generator column material. Appl Radiat Isot 2019; 149:206-213. [PMID: 31078965 DOI: 10.1016/j.apradiso.2019.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/15/2019] [Accepted: 04/13/2019] [Indexed: 10/27/2022]
Abstract
A durable and ready to use 68Ge-68Ga generator column material is required for its routine use in radiopharmaceutical procedures. The present work comprises preliminary studies for development and evaluation of chitosan-TiO2 based microsphere (C-TOM) composite towards its competence as a column material. The batch uptake studies showed higher distribution coefficients for 68Ge vis-à-vis 68Ga in the complete concentration range of HCl examined (0.01-1 mol.L-1). Furthermore, C-TOM showed enduring physical and chemical stability in 0.01 mol.L-1 HCl with persistent 68Ga elution profiles (>95%) and negligible 68Ge breakthrough (2 × 10-4%) for the preliminary evaluation period of ∼2 months. Overall, the studies indicated that, 68Ga with high radionuclidic purity (≥99.99%) can be eluted routinely in a small volume (∼1.5 mL) of 0.01 mol.L-1 HCl proving its potentials as a novel solid phase extractant for 68Ge/68Ge generator system.
Collapse
Affiliation(s)
- Chirag K Vyas
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Jun Young Lee
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Min Goo Hur
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Seung Dae Yang
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Young Bae Kong
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Eun Je Lee
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea
| | - Jeong Hoon Park
- Radiation Instrumentation Research Division, Korea Atomic Energy Research Institute, Jeongeup Si, Jeollabuk Do - 56212, Republic of Korea.
| |
Collapse
|
34
|
González-Flores E, Serrano R, Sevilla I, Viúdez A, Barriuso J, Benavent M, Capdevila J, Jimenez-Fonseca P, López C, Garcia-Carbonero R. SEOM clinical guidelines for the diagnosis and treatment of gastroenteropancreatic and bronchial neuroendocrine neoplasms (NENs) (2018). Clin Transl Oncol 2019; 21:55-63. [PMID: 30535553 PMCID: PMC6339660 DOI: 10.1007/s12094-018-1980-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
Abstract
NENs are a heterogeneous family of tumors of challenging diagnosis and clinical management. Their incidence and prevalence continue to rise across all sites, stages and grades. Although improved diagnostic techniques have led to earlier detection and stage migration, the improved prognosis documented over time for advanced gastrointestinal and pancreatic neuroendocrine tumors also reflect improvements in therapy. The aim of this guideline is to update practical recommendations for the diagnosis and treatment of gastroenteropancreatic and lung NENs. Diagnostic procedures, histological classification and therapeutic options are briefly discussed, including surgery, liver-directed therapy, peptide receptor radionuclide therapy, and systemic hormonal, cytotoxic or targeted therapy, and treatment algorithms are provided.
Collapse
Affiliation(s)
- E. González-Flores
- Department of Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - R. Serrano
- Department of Medical Oncology, Hospital Reina Sofía, Córdoba, IMIBIC, CIBERONC, Córdoba, Spain
| | - I. Sevilla
- Department of Medical Oncology, Instituto de Investigaciones Biomédicas de Málaga (IBIMA)/Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, Málaga, Spain
| | - A. Viúdez
- Department of Medical Oncology, Complejo Hospitalario de Navarra (CHN), OncobionaTras Unit, Navarrabiomed, IdiSNA, Pamplona, Spain
| | - J. Barriuso
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - M. Benavent
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina, Seville, Spain
| | - J. Capdevila
- Department of Medical Oncology, Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VIHO), Barcelona, Spain
| | - P. Jimenez-Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - C. López
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - R. Garcia-Carbonero
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, IIS imas12, UCM, CNIO, CIBERONC, Av. de Córdoba, s/n, 28041 Madrid, Spain
| |
Collapse
|
35
|
Franco Machado J, Silva RD, Melo R, G Correia JD. Less Exploited GPCRs in Precision Medicine: Targets for Molecular Imaging and Theranostics. Molecules 2018; 24:E49. [PMID: 30583594 PMCID: PMC6337414 DOI: 10.3390/molecules24010049] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/07/2018] [Accepted: 12/09/2018] [Indexed: 12/18/2022] Open
Abstract
Precision medicine relies on individually tailored therapeutic intervention taking into account individual variability. It is strongly dependent on the availability of target-specific drugs and/or imaging agents that recognize molecular targets and patient-specific disease mechanisms. The most sensitive molecular imaging modalities, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET), rely on the interaction between an imaging radioprobe and a target. Moreover, the use of target-specific molecular tools for both diagnostics and therapy, theranostic agents, represent an established methodology in nuclear medicine that is assuming an increasingly important role in precision medicine. The design of innovative imaging and/or theranostic agents is key for further accomplishments in the field. G-protein-coupled receptors (GPCRs), apart from being highly relevant drug targets, have also been largely exploited as molecular targets for non-invasive imaging and/or systemic radiotherapy of various diseases. Herein, we will discuss recent efforts towards the development of innovative imaging and/or theranostic agents targeting selected emergent GPCRs, namely the Frizzled receptor (FZD), Ghrelin receptor (GHSR-1a), G protein-coupled estrogen receptor (GPER), and Sphingosine-1-phosphate receptor (S1PR). The pharmacological and clinical relevance will be highlighted, giving particular attention to the studies on the synthesis and characterization of targeted molecular imaging agents, biological evaluation, and potential clinical applications in oncology and non-oncology diseases. Whenever relevant, supporting computational studies will be also discussed.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal.
| | - Rúben D Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - Rita Melo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
- Center for Neuroscience and Cell Biology; Rua Larga, Faculdade de Medicina, Polo I, 1ºandar, Universidade de Coimbra, 3004-504 Coimbra, Portugal.
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| |
Collapse
|
36
|
Werner RA, Weich A, Kircher M, Solnes LB, Javadi MS, Higuchi T, Buck AK, Pomper MG, Rowe SP, Lapa C. The theranostic promise for Neuroendocrine Tumors in the late 2010s - Where do we stand, where do we go? Theranostics 2018; 8:6088-6100. [PMID: 30613284 PMCID: PMC6299695 DOI: 10.7150/thno.30357] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
More than 25 years after the first peptide receptor radionuclide therapy (PRRT), the concept of somatostatin receptor (SSTR)-directed imaging and therapy for neuroendocrine tumors (NET) is seeing rapidly increasing use. To maximize the full potential of its theranostic promise, efforts in recent years have expanded recommendations in current guidelines and included the evaluation of novel theranostic radiotracers for imaging and treatment of NET. Moreover, the introduction of standardized reporting framework systems may harmonize PET reading, address pitfalls in interpreting SSTR-PET/CT scans and guide the treating physician in selecting PRRT candidates. Notably, the concept of PRRT has also been applied beyond oncology, e.g. for treatment of inflammatory conditions like sarcoidosis. Future perspectives may include the efficacy evaluation of PRRT compared to other common treatment options for NET, novel strategies for closer monitoring of potential side effects, the introduction of novel radiotracers with beneficial pharmacodynamic and kinetic properties or the use of supervised machine learning approaches for outcome prediction. This article reviews how the SSTR-directed theranostic concept is currently applied and also reflects on recent developments that hold promise for the future of theranostics in this context.
Collapse
Affiliation(s)
- Rudolf A. Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Alexander Weich
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Germany
| | - Malte Kircher
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Lilja B. Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehrbod S. Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- Department of Bio Medical Imaging, National Cardiovascular and Cerebral Research Center, Suita, Japan
| | - Andreas K. Buck
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Constantin Lapa
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| |
Collapse
|
37
|
Pauwels E, Cleeren F, Bormans G, Deroose CM. Somatostatin receptor PET ligands - the next generation for clinical practice. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2018; 8:311-331. [PMID: 30510849 PMCID: PMC6261874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/04/2018] [Indexed: 06/09/2023]
Abstract
Somatostatin receptors (SSTRs) are variably expressed by a variety of malignancies. Using radiolabeled somatostatin analogs (SSAs), the presence of SSTRs on tumor cells may be exploited for molecular imaging and for peptide receptor radionuclide therapy. 111In-DTPA-octreotide has long been the standard in SSTR scintigraphy. A major leap forward was the introduction of gallium-68 labeled SSAs for positron emission tomography (PET) offering improved sensitivity. Tracers currently in clinical use are 68Ga-DOTA-Tyr3-octreotide (68Ga-DOTATOC), 68Ga-DOTA-Tyr3-octreotate (68Ga-DOTATATE) and 68Ga-DOTA-1-NaI3-octreotide (68Ga-DOTANOC), collectively referred to as 68Ga-DOTA-peptides. 68Ga-DOTA-peptide PET has superseded 111In-DTPA-octreotide scintigraphy as the modality of choice for SSTR imaging. However, implementation of 68Ga-DOTA-peptides in routine clinical practice is often limited by practical, economical and regulatory factors related to the use of the current generation of 68Ge/68Ga generators. Centralized production and distribution is challenging due to the low production yield and relatively short half-life of gallium-68. Furthermore, gallium-68 has a relatively long positron range, compromising spatial resolution on modern PET cameras. Therefore, possibilities of using other PET radionuclides are being explored. On the other hand, new developments in SSTR PET ligands are strongly driven by the need for improved lesion targeting, especially for tumors with low SSTR expression. This may be achieved by using peptide vectors having a higher affinity for the SSTR or a broader affinity profile for the different receptor subtypes or by using compounds recognizing more binding sites, such as SSTR antagonists. This review gives an overview of recent developments leading to the next generation of clinical PET tracers for SSTR imaging.
Collapse
Affiliation(s)
- Elin Pauwels
- Nuclear Medicine, University Hospitals LeuvenLeuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU LeuvenLeuven, Belgium
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU LeuvenLeuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU LeuvenLeuven, Belgium
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals LeuvenLeuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU LeuvenLeuven, Belgium
| |
Collapse
|
38
|
Werner RA, Chen X, Rowe SP, Lapa C, Javadi MS, Higuchi T. Moving into the next era of PET myocardial perfusion imaging: introduction of novel 18F-labeled tracers. Int J Cardiovasc Imaging 2018; 35:569-577. [PMID: 30334228 PMCID: PMC6454078 DOI: 10.1007/s10554-018-1469-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/12/2018] [Indexed: 01/15/2023]
Abstract
The heart failure epidemic continues to rise with coronary artery disease as one of its main causes. Novel concepts for risk stratification to guide the referring cardiologist towards revascularization procedures are of significant value. Myocardial perfusion imaging using single-photon emission computed tomography (SPECT) agents has demonstrated high accuracy for the detection of clinically relevant stenoses. With positron emission tomography (PET) becoming more widely available, mainly due to its diagnostic performance in oncology, perfusion imaging with that modality is more practical than in the past and overcomes existing limitations of SPECT MPI. Advantages of PET include more reliable quantification of absolute myocardial blood flow, the routine use of computed tomography for attenuation correction, a higher spatiotemporal resolution and a higher count sensitivity. Current PET radiotracers such as rubidium-82 (half-life, 76 s), oxygen-15 water (2 min) or nitrogen-13 ammonia (10 min) are labeled with radionuclides with very short half-lives, necessitating that stress imaging is performed under pharmacological vasodilator stress instead of exercise testing. However, with the introduction of novel 18F-labeled MPI PET radiotracers (half-life, 110 min), the intrinsic advantages of PET can be combined with exercise testing. Additional advantages of those radiotracers include, but are not limited to: potentially improved cost-effectiveness due to the use of pre-existing delivery systems and superior imaging qualities, mainly due to the shortest positron range among available PET MPI probes. In the present review, widely used PET MPI radiotracers will be reviewed and potential novel 18F-labeled perfusion radiotracers will be discussed.
Collapse
Affiliation(s)
- Rudolf A Werner
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Nuclear Medicine, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany
| | - Xinyu Chen
- Department of Nuclear Medicine, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Constantin Lapa
- Department of Nuclear Medicine, University of Wuerzburg, Wuerzburg, Germany
| | - Mehrbod S Javadi
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University of Wuerzburg, Wuerzburg, Germany. .,Comprehensive Heart Failure Center, University of Wuerzburg, Oberduerrbacher Strasse 6, 97080, Wuerzburg, Germany. .,Department of Biomedical Imaging, National Cardiovascular and Cerebral Center, Suita, Japan.
| |
Collapse
|
39
|
Werner RA, Bundschuh RA, Bundschuh L, Javadi MS, Higuchi T, Weich A, Sheikhbahaei S, Pienta KJ, Buck AK, Pomper MG, Gorin MA, Lapa C, Rowe SP. Molecular imaging reporting and data systems (MI-RADS): a generalizable framework for targeted radiotracers with theranostic implications. Ann Nucl Med 2018; 32:512-522. [PMID: 30109562 PMCID: PMC6182628 DOI: 10.1007/s12149-018-1291-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 01/15/2023]
Abstract
Both prostate-specific membrane antigen (PSMA)- and somatostatin receptor (SSTR)-targeted positron emission tomography (PET)-based imaging agents for prostate carcinoma and neuroendocrine tumors, respectively, are seeing rapidly expanding use. In addition to diagnostic applications, both classes of radiotracers can be used to triage patients for theranostic endoradiotherapy. While interpreting PSMA- or SSTR-targeted PET/computed tomography (CT) scans, the reader has to be aware of certain pitfalls. Adding to the complexity of the interpretation of those imaging agents, both normal biodistribution, and also false-positive and -negative findings differ between PSMA- and SSTR-targeted PET radiotracers. Herein summarized under the umbrella term molecular imaging reporting and data systems (MI-RADS), two novel RADS classifications for PSMA- and SSTR-targeted PET imaging are described (PSMA- and SSTR-RADS). Notably, PSMA- and SSTR-RADS are structured in a reciprocal fashion, i.e., if the reader is familiar with one system, the other system can readily be applied, as well. In the present review, we will discuss the most common pitfalls on PSMA- and SSTR-targeted PET/CT, briefly introduce PSMA- and SSTR-RADS, and define a potential future role of the umbrella framework MI-RADS compared to other classification systems.
Collapse
Affiliation(s)
- Rudolf A Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA.,Department of Nuclear Medicine, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,European Neuroendocrine Tumor Society (ENETS), Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Würzburg, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Lena Bundschuh
- Department of Nuclear Medicine, University Medical Center Bonn, Bonn, Germany
| | - Mehrbod S Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,Department of Bio Medical Imaging, National Cardiovascular and Cerebral Research Center, Suita, Japan
| | - Alexander Weich
- European Neuroendocrine Tumor Society (ENETS), Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Würzburg, Germany.,Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Würzburg, Germany
| | - Sara Sheikhbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA
| | - Kenneth J Pienta
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas K Buck
- Department of Nuclear Medicine, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,European Neuroendocrine Tumor Society (ENETS), Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Würzburg, Germany
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA
| | - Michael A Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA.,Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Constantin Lapa
- Department of Nuclear Medicine, Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,European Neuroendocrine Tumor Society (ENETS), Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Würzburg, Germany
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 601 N. Caroline St., Baltimore, MD, 21287, USA. .,Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
Stéen EJL, Edem PE, Nørregaard K, Jørgensen JT, Shalgunov V, Kjaer A, Herth MM. Pretargeting in nuclear imaging and radionuclide therapy: Improving efficacy of theranostics and nanomedicines. Biomaterials 2018; 179:209-245. [PMID: 30007471 DOI: 10.1016/j.biomaterials.2018.06.021] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 01/18/2023]
Abstract
Pretargeted nuclear imaging and radiotherapy have recently attracted increasing attention for diagnosis and treatment of cancer with nanomedicines. This is because it conceptually offers better imaging contrast and therapeutic efficiency while reducing the dose to radiosensitive tissues compared to conventional strategies. In conventional imaging and radiotherapy, a directly radiolabeled nano-sized vector is administered and allowed to accumulate in the tumor, typically on a timescale of several days. In contrast, pretargeting is based on a two-step approach. First, a tumor-accumulating vector carrying a tag is administered followed by injection of a fast clearing radiolabeled agent that rapidly recognizes the tag of the tumor-bound vector in vivo. Therefore, pretargeting circumvents the use of long-lived radionuclides that is a necessity for sufficient tumor accumulation and target-to-background ratios using conventional approaches. In this review, we give an overview of recent advances in pretargeted imaging strategies. We will critically reflect on the advantages and disadvantages of current state-of-the-art conventional imaging approaches and compare them to pretargeted strategies. We will discuss the pretargeted imaging concept and the involved chemistry. Finally, we will discuss the steps forward in respect to clinical translation, and how pretargeted strategies could be applied to improve state-of-the-art radiotherapeutic approaches.
Collapse
Affiliation(s)
- E Johanna L Stéen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Patricia E Edem
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Kamilla Nørregaard
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2100 Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
41
|
Nicolas GP, Beykan S, Bouterfa H, Kaufmann J, Bauman A, Lassmann M, Reubi JC, Rivier JE, Maecke HR, Fani M, Wild D. Safety, Biodistribution, and Radiation Dosimetry of 68Ga-OPS202 in Patients with Gastroenteropancreatic Neuroendocrine Tumors: A Prospective Phase I Imaging Study. J Nucl Med 2017; 59:909-914. [DOI: 10.2967/jnumed.117.199737] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/30/2017] [Indexed: 12/24/2022] Open
|
42
|
Development of a radiolabeled caninized anti-EGFR antibody for comparative oncology trials. Oncotarget 2017; 8:83128-83141. [PMID: 29137329 PMCID: PMC5669955 DOI: 10.18632/oncotarget.20914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Due to large homology of human and canine EGFR, dogs suffering from spontaneous EGFR+ cancer can be considered as ideal translational models. Thereby, novel immunotherapeutic compounds can be developed for both human and veterinary patients. This study describes the radiolabeling of a canine anti-EGFR IgG antibody (can225IgG) with potential diagnostic and therapeutic value in comparative clinical settings. Can225IgG was functionalized with DTPA for subsequent chelation with the radionuclide 99mTc. Successful coupling of 10 DTPA molecules per antibody on average was proven by significant mass increase in MALDI-TOF spectroscopy, gel electrophoresis and immunoblots. Following functionalization and radiolabeling, 99mTc-DTPA-can225IgG fully retained its binding capacity towards human and canine EGFR in flow cytometry, immuno- and radioblots, and autoradiography. The affinity of radiolabeled can225IgG was determined to KD 0.8 ±0.0031 nM in a real-time kinetics assay on canine carcinoma cells by a competition binding technique. Stability tests of the radiolabeled compound identified TRIS buffered saline as the ideal formulation for short-term storage with 87.11 ±6.04% intact compound being still detected 60 minutes post radiolabeling. High stability, specificity and EGFR binding affinity pinpoint towards 99mTc-radiolabeled can225IgG antibody as an ideal lead compound for the first proof-of-concept diagnostic and therapeutic applications in canine cancer patients.
Collapse
|
43
|
Fani M, Peitl PK, Velikyan I. Current Status of Radiopharmaceuticals for the Theranostics of Neuroendocrine Neoplasms. Pharmaceuticals (Basel) 2017; 10:E30. [PMID: 28295000 PMCID: PMC5374434 DOI: 10.3390/ph10010030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
Nuclear medicine plays a pivotal role in the management of patients affected by neuroendocrine neoplasms (NENs). Radiolabeled somatostatin receptor analogs are by far the most advanced radiopharmaceuticals for diagnosis and therapy (radiotheranostics) of NENs. Their clinical success emerged receptor-targeted radiolabeled peptides as an important class of radiopharmaceuticals and it paved the way for the investigation of other radioligand-receptor systems. Besides the somatostatin receptors (sstr), other receptors have also been linked to NENs and quite a number of potential radiolabeled peptides have been derived from them. The Glucagon-Like Peptide-1 Receptor (GLP-1R) is highly expressed in benign insulinomas, the Cholecystokinin 2 (CCK2)/Gastrin receptor is expressed in different NENs, in particular medullary thyroid cancer, and the Glucose-dependent Insulinotropic Polypeptide (GIP) receptor was found to be expressed in gastrointestinal and bronchial NENs, where interestingly, it is present in most of the sstr-negative and GLP-1R-negative NENs. Also in the field of sstr targeting new discoveries brought into light an alternative approach with the use of radiolabeled somatostatin receptor antagonists, instead of the clinically used agonists. The purpose of this review is to present the current status and the most innovative strategies for the diagnosis and treatment (theranostics) of neuroendocrine neoplasms using a cadre of radiolabeled regulatory peptides targeting their receptors.
Collapse
Affiliation(s)
- Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital of Basel, 4031 Basel, Switzerland.
| | - Petra Kolenc Peitl
- Department of Nuclear Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia.
| | - Irina Velikyan
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden.
| |
Collapse
|
44
|
Pham THN, Lengkeek NA, Greguric I, Kim BJ, Pellegrini PA, Bickley SA, Tanudji MR, Jones SK, Hawkett BS, Pham BTT. Tunable and noncytotoxic PET/SPECT-MRI multimodality imaging probes using colloidally stable ligand-free superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2017; 12:899-909. [PMID: 28184160 PMCID: PMC5291326 DOI: 10.2147/ijn.s127171] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Physiologically stable multimodality imaging probes for positron emission tomography/single-photon emission computed tomography (PET/SPECT)-magnetic resonance imaging (MRI) were synthesized using the superparamagnetic maghemite iron oxide (γ-Fe2O3) nanoparticles (SPIONs). The SPIONs were sterically stabilized with a finely tuned mixture of diblock copolymers with either methoxypolyethylene glycol (MPEG) or primary amine NH2 end groups. The radioisotope for PET or SPECT imaging was incorporated with the SPIONs at high temperature. 57Co2+ ions with a long half-life of 270.9 days were used as a model for the radiotracer to study the kinetics of radiolabeling, characterization, and the stability of the radiolabeled SPIONs. Radioactive 67Ga3+ and Cu2+-labeled SPIONs were also produced successfully using the optimized conditions from the 57Co2+-labeling process. No free radioisotopes were detected in the aqueous phase for the radiolabeled SPIONs 1 week after dispersion in phosphate-buffered saline (PBS). All labeled SPIONs were not only well dispersed and stable under physiological conditions but also noncytotoxic in vitro. The ability to design and produce physiologically stable radiolabeled magnetic nanoparticles with a finely controlled number of functionalizable end groups on the SPIONs enables the generation of a desirable and biologically compatible multimodality PET/SPECT-MRI agent on a single T2 contrast MRI probe.
Collapse
Affiliation(s)
- TH Nguyen Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Nigel A Lengkeek
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | - Ivan Greguric
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | - Byung J Kim
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Paul A Pellegrini
- Radioisotopes and Radiotracers, NSTLI, Australian Nuclear Science and Technology Organisation, Sydney
| | | | | | | | - Brian S Hawkett
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| | - Binh TT Pham
- Key Centre for Polymers and Colloids, School of Chemistry, University of Sydney
| |
Collapse
|
45
|
Gomez JDC, Hagenbach A, Gerling-Driessen UIM, Koksch B, Beindorff N, Brenner W, Abram U. Thiourea derivatives as chelating agents for bioconjugation of rhenium and technetium. Dalton Trans 2017; 46:14602-14611. [DOI: 10.1039/c7dt01834g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A99mTc complex with a tetradentate thiocarbamoylbenzamidine group was used for the conjugation of angiotensin-II. The resulting bioconjugate is stablein vivoandin vitro.
Collapse
Affiliation(s)
- J. D. Castillo Gomez
- Freie Universität Berlin
- Institute of Chemistry and Biochemistry
- D-14195 Berlin
- Germany
| | - A. Hagenbach
- Freie Universität Berlin
- Institute of Chemistry and Biochemistry
- D-14195 Berlin
- Germany
| | | | - B. Koksch
- Freie Universität Berlin
- Institute of Chemistry and Biochemistry
- D-14195 Berlin
- Germany
| | - N. Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC)
- D-13353 Berlin
- Germany
| | - W. Brenner
- Department of Nuclear Medicine
- Charité
- Campus Virchow Klinikum
- D-13353 Berlin
- Germany
| | - U. Abram
- Freie Universität Berlin
- Institute of Chemistry and Biochemistry
- D-14195 Berlin
- Germany
| |
Collapse
|
46
|
Emoto MC, Sato S, Fujii HG. Development of nitroxide-based theranostic compounds that act both as anti-inflammatory drugs and brain redox imaging probes in MRI. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2016; 54:705-711. [PMID: 26990690 DOI: 10.1002/mrc.4431] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/05/2016] [Accepted: 02/21/2016] [Indexed: 06/05/2023]
Abstract
Theranostic probes provide both therapeutic and diagnostic imaging capabilities in one molecule and show significant promise for use in magnetic resonance imaging (MRI) examinations. The present study describes for the first time the synthesis and utility of nitroxide-based contrast agents exhibiting a nonsteroidal anti-inflammatory drug effect. The target theranostic probes were prepared by connecting the carboxyl group of ibuprofen or ketoprofen to the hydroxyl group of 3-hydroxymethyl-2,2,5,5-tetramethylprrolidine-1-oxyl by a condensation reaction in the presence of dicyclohexylcarbodiimide and 4-dimethylaminopyridine in dichloromethane. MRI of mouse heads after administration of either synthesized theranostic probe indicated that the probes enter the brain by passing through the blood-brain barrier (BBB), resulting in T1 contrast enhancement in mouse brain. This enhancement persisted for the duration of the half-life of about 40 min, which is longer than that obtained by most of pyrrolidine nitroxide molecules. The therapeutic capacities of these theranostic probes were examined using a lipopolysaccharide (LPS)-induced brain inflammation model. The production of nitric oxide, an inflammation marker in septic mouse brain induced by LPS, was remarkably inhibited by the addition of either synthesized probe, indicating that they also act as anti-inflammatory drugs. The present results indicate that nitroxide-based theranostic probes act as both BBB-permeable redox-sensitive contrast agents and as an anti-inflammatory drug in septic mouse brain. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Miho C Emoto
- Center for Medical Education, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Shingo Sato
- Graduate School of Science and Engineering, Yamagata University, Yonezawa, Yamagata, Japan
| | - Hirotada G Fujii
- Center for Medical Education, Sapporo Medical University, Sapporo, Hokkaido, Japan
| |
Collapse
|
47
|
Pfob CH, Ziegler S, Graner FP, Köhner M, Schachoff S, Blechert B, Wester HJ, Scheidhauer K, Schwaiger M, Maurer T, Eiber M. Biodistribution and radiation dosimetry of (68)Ga-PSMA HBED CC-a PSMA specific probe for PET imaging of prostate cancer. Eur J Nucl Med Mol Imaging 2016; 43:1962-70. [PMID: 27207281 DOI: 10.1007/s00259-016-3424-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 05/11/2016] [Indexed: 12/18/2022]
Abstract
PURPOSE Positron emission tomography (PET) agents targeting the prostate-specific membrane antigen (PSMA) are currently under broad clinical and scientific investigation. (68)Ga-PSMA HBED-CC constitutes the first (68)Ga-labelled PSMA-inhibitor and has evolved as a promising agent for imaging PSMA expression in vivo. The aim of this study was to evaluate the whole-body distribution and radiation dosimetry of this new probe. METHODS Five patients with a history or high suspicion of prostate cancer were injected intravenously with a mean of 139.8 ± 13.7 MBq of (68)Ga-PSMA HBED-CC (range 120-158 MBq). Four static skull to mid-thigh scans using a whole-body fully integrated PET/MR-system were performed 10 min, 60 min, 130 min, and 175 min after the tracer injection. Time-dependent changes of the injected activity per organ were determined. Mean organ-absorbed doses and effective doses (ED) were calculated using OLINDA/EXM. RESULTS Injection of a standard activity of 150 MBq (68)Ga-PSMA HBED-CC resulted in a median effective dose of 2.37 mSv (Range 1.08E-02 - 2.46E-02 mSv/MBq). The urinary bladder wall (median absorbed dose 1.64E-01 mGv/MBq; range 8.76E-02 - 2.91E-01 mGv/MBq) was the critical organ, followed by the kidneys (median absorbed dose 1.21E-01 mGv/MBq; range 7.16E-02 - 1.75E-01), spleen (median absorbed dose 4.13E-02 mGv/MBq; range 1.57E-02 - 7.32E-02 mGv/MBq) and liver (median absorbed dose 2.07E-02 mGv/MBq; range 1.80E-02 - 2.57E-02 mGv/MBq). No drug-related pharmacological effects occurred. CONCLUSION The use of (68)Ga-PSMA HBED-CC results in a relatively low radiation exposure, delivering organ doses that are comparable to those of other (68)Ga-labelled PSMA-inhibitors used for PET-imaging. Total effective dose is lower than for other PET-agents used for prostate cancer imaging (e.g. (11)C- and (18)F-Choline).
Collapse
Affiliation(s)
- Christian H Pfob
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany.
| | - Sibylle Ziegler
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Frank Philipp Graner
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Markus Köhner
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Sylvia Schachoff
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Birgit Blechert
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Department Chemie, Technische Universität München, Walther-Meissner-Str. 3, 85748, Garching, Germany
| | - Klemens Scheidhauer
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| | - Tobias Maurer
- Department of Urology, Technische Universität München, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar; Ismaningerstrasse 22, 81675, Munich, Germany
| |
Collapse
|
48
|
de Galiza Barbosa F, Delso G, Ter Voert EEGW, Huellner MW, Herrmann K, Veit-Haibach P. Multi-technique hybrid imaging in PET/CT and PET/MR: what does the future hold? Clin Radiol 2016; 71:660-72. [PMID: 27108800 DOI: 10.1016/j.crad.2016.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/11/2016] [Accepted: 03/22/2016] [Indexed: 12/19/2022]
Abstract
Integrated positron-emission tomography and computed tomography (PET/CT) is one of the most important imaging techniques to have emerged in oncological practice in the last decade. Hybrid imaging, in general, remains a rapidly growing field, not only in developing countries, but also in western industrialised healthcare systems. A great deal of technological development and research is focused on improving hybrid imaging technology further and introducing new techniques, e.g., integrated PET and magnetic resonance imaging (PET/MRI). Additionally, there are several new PET tracers on the horizon, which have the potential to broaden clinical applications in hybrid imaging for diagnosis as well as therapy. This article aims to highlight some of the major technical and clinical advances that are currently taking place in PET/CT and PET/MRI that will potentially maintain the position of hybrid techniques at the forefront of medical imaging technologies.
Collapse
Affiliation(s)
- F de Galiza Barbosa
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland
| | - G Delso
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; GE Healthcare, Waukesha, WI, USA
| | - E E G W Ter Voert
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland
| | - M W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Department of Neuroradiology, University Hospital Zurich, Switzerland
| | - K Herrmann
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, United States; Department of Nuclear Medicine, Universitätsklinikum Würzburg, Oberdürrbacher, Str. 6, Würzburg, Germany
| | - P Veit-Haibach
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Department of Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland.
| |
Collapse
|
49
|
Spick C, Herrmann K, Czernin J. 18F-FDG PET/CT and PET/MRI Perform Equally Well in Cancer: Evidence from Studies on More Than 2,300 Patients. J Nucl Med 2016; 57:420-30. [PMID: 26742709 PMCID: PMC5003572 DOI: 10.2967/jnumed.115.158808] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/05/2016] [Indexed: 12/31/2022] Open
Abstract
(18)F-FDG PET/CT has become the reference standard in oncologic imaging against which the performance of other imaging modalities is measured. The promise of PET/MRI includes multiparametric imaging to further improve diagnosis and phenotyping of cancer. Rather than focusing on these capabilities, many investigators have examined whether (18)F-FDG PET combined with mostly anatomic MRI improves cancer staging and restaging. After a description of PET/MRI scanner designs and a discussion of technical and operational issues, we review the available literature to determine whether cancer assessments are improved with PET/MRI. The available data show that PET/MRI is feasible and performs as well as PET/CT in most types of cancer. Diagnostic advantages may be achievable in prostate cancer and in bone metastases, whereas disadvantages exist in lung nodule assessments. We conclude that (18)F-FDG PET/MRI and PET/CT provide comparable diagnostic information when MRI is used simply to provide the anatomic framework. Thus, PET/MRI could be used in lieu of PET/CT if this approach becomes economically viable and if reasonable workflows can be established. Future studies should explore the multiparametric potential of MRI.
Collapse
Affiliation(s)
- Claudio Spick
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California; and
| | - Ken Herrmann
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California; and Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Czernin
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California; and
| |
Collapse
|
50
|
Ellison PA, Barnhart TE, Chen F, Hong H, Zhang Y, Theuer CP, Cai W, Nickles RJ, DeJesus OT. High Yield Production and Radiochemical Isolation of Isotopically Pure Arsenic-72 and Novel Radioarsenic Labeling Strategies for the Development of Theranostic Radiopharmaceuticals. Bioconjug Chem 2015; 27:179-88. [PMID: 26646989 DOI: 10.1021/acs.bioconjchem.5b00592] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Radioisotopes of arsenic are of considerable interest to the field of nuclear medicine with unique nuclear and chemical properties making them well-suited for use in novel theranostic radiopharmaceuticals. However, progress must still be made in the production of isotopically pure radioarsenic and in its stable conjugation to biological targeting vectors. This work presents the production and irradiation of isotopically enriched (72)Ge(m) discs in an irrigation-cooled target system allowing for the production of isotopically pure (72)As with capability on the order of 10 GBq. A radiochemical separation procedure isolated the reactive trivalent radioarsenic in a small volume buffered aqueous solution, while reclaiming (72)Ge target material. The direct thiol-labeling of a monoclonal antibody resulted in a conjugate exhibiting exceptionally poor in vivo stability in a mouse model. This prompted further investigations to alternative radioarsenic labeling strategies, including the labeling of the dithiol-containing chelator dihydrolipoic acid, and thiol-modified mesoporous silica nanoparticles (MSN-SH). Radioarsenic-labeled MSN-SH showed exceptional in vivo stability toward dearsenylation.
Collapse
Affiliation(s)
| | | | | | | | | | - Charles P Theuer
- TRACON Pharmaceuticals, Inc. , San Diego, California 92122, United States
| | - Weibo Cai
- Carbone Cancer Center and Materials Science Program, University of Wisconsin , Madison, Wisconsin 53706, United States
| | | | | |
Collapse
|