1
|
Ebner R, Lohse A, Fabritius MP, Rübenthaler J, Wängler C, Wängler B, Schirrmacher R, Völter F, Schmid HP, Unterrainer LM, Öcal O, Hinterberger A, Spitzweg C, Auernhammer CJ, Geyer T, Ricke J, Bartenstein P, Holzgreve A, Grawe F. Validation of the standardization framework SSTR-RADS 1.0 for neuroendocrine tumors using the novel SSTR‑targeting peptide [ 18F]SiTATE. Eur Radiol 2024; 34:7222-7232. [PMID: 38769164 PMCID: PMC11519286 DOI: 10.1007/s00330-024-10788-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/27/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVES Somatostatin receptor positron emission tomography/computed tomography (SSTR-PET/CT) using [68Ga]-labeled tracers is a widely used imaging modality for neuroendocrine tumors (NET). Recently, [18F]SiTATE, a SiFAlin tagged [Tyr3]-octreotate (TATE) PET tracer, has shown great potential due to favorable clinical characteristics. We aimed to evaluate the reproducibility of Somatostatin Receptor-Reporting and Data System 1.0 (SSTR-RADS 1.0) for structured interpretation and treatment planning of NET using [18F]SiTATE. METHODS Four readers assessed [18F]SiTATE-PET/CT of 95 patients according to the SSTR-RADS 1.0 criteria at two different time points. Each reader evaluated up to five target lesions per scan. The overall scan score and the decision on peptide receptor radionuclide therapy (PRRT) were considered. Inter- and intra-reader agreement was determined using the intraclass correlation coefficient (ICC). RESULTS The ICC analysis on the inter-reader agreement using SSTR-RADS 1.0 for identical target lesions (ICC ≥ 85%), overall scan score (ICC ≥ 90%), and the decision to recommend PRRT (ICC ≥ 85%) showed excellent agreement. However, significant differences were observed in recommending PRRT among experienced readers (ER) (p = 0.020) and inexperienced readers (IR) (p = 0.004). Compartment-based analysis demonstrated good to excellent inter-reader agreement for most organs (ICC ≥ 74%), except for lymph nodes (ICC ≥ 53%). CONCLUSION SSTR-RADS 1.0 represents a highly reproducible and consistent framework system for stratifying SSTR-targeted PET/CT scans, even using the novel SSTR-ligand [18F]SiTATE. Some inter-reader variability was observed regarding the evaluation of uptake intensity prior to PRRT as well as compartment scoring of lymph nodes, indicating that those categories require special attention during further clinical validation and might be refined in a future SSTR-RADS version 1.1. CLINICAL RELEVANCE STATEMENT SSTR-RADS 1.0 is a consistent framework for categorizing somatostatin receptor-targeted PET/CT scans when using [18F]SiTATE. The framework serves as a valuable tool for facilitating and improving the management of patients with NET. KEY POINTS SSTR-RADS 1.0 is a valuable tool for managing patients with NET. SSTR-RADS 1.0 categorizes patients with showing strong agreement across diverse reader expertise. As an alternative to [68Ga]-labeled PET/CT in neuroendocrine tumor imaging, SSTR-RADS 1.0 reliably classifies [18F]SiTATE-PET/CT.
Collapse
Affiliation(s)
- R Ebner
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - A Lohse
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - M P Fabritius
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - J Rübenthaler
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
| | - C Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - B Wängler
- Biomedical Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
- Molecular Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - R Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Canada
| | - F Völter
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - H P Schmid
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - L M Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - O Öcal
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - A Hinterberger
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Heidelberg, Germany
| | - C Spitzweg
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - C J Auernhammer
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - T Geyer
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - J Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
| | - P Bartenstein
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - A Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - F Grawe
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Heidelberg, Germany
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
2
|
Şahin R, Baloğlu MC, Ergül N, Çermik TF, Arslan E. 68 Ga-DOTA-FAPI-46 PET/CT Imaging for Restaging in a Patient With Metastatic Pheochromocytoma : Comparison With 68 Ga-DOTA-TATE PET/CT. Clin Nucl Med 2024; 49:e622-e624. [PMID: 39365092 DOI: 10.1097/rlu.0000000000005481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
ABSTRACT Pheochromocytomas (PHEOs) are neural crest-derived tumors originating from the chromaffin cells of the adrenal medulla and were recognized as one of the subtypes of paragangliomas by the World Health Organization in 2022. 68 Ga-labeled somatostatin analog ( 68 Ga-DOTA-NOC, 68 Ga-DOTA-TOC, and 68 Ga-DOTA-TATE) PET imaging has shown significant performance compared with 123 MIBG scintigraphy in the diagnosis of paragangliomas. It is now known that fibroblast activation protein (FAP) is overexpressed by various cancer-associated fibroblasts, including PHEOs. We would like to present the findings of 68 Ga-DOTA-FAPI-46 PET/CT and 68 Ga-DOTA-TATE PET/CT imaging performed for restaging on a 42-year-old man diagnosed with metastatic PHEO.
Collapse
Affiliation(s)
- Rahime Şahin
- From the Clinic of Nuclear Medicine, Istanbul Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
3
|
Ebner R, Sheikh GT, Brendel M, Ricke J, Cyran CC. ESR Essentials: role of PET/CT in neuroendocrine tumors-practice recommendations by the European Society for Hybrid, Molecular and Translational Imaging. Eur Radiol 2024:10.1007/s00330-024-11095-7. [PMID: 39387873 DOI: 10.1007/s00330-024-11095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024]
Abstract
Neuroendocrine neoplasms (NEN) originate from the secretory cells of the neuroendocrine system, with the majority arising in the gastrointestinal tract and pancreas. Given the heterogeneity in the biological behavior and morphological differentiation of these tumors, advanced imaging techniques are crucial for supporting the suspected diagnosis, accurate staging, and monitoring therapy. As most well-differentiated NEN demonstrate overexpression of somatostatin receptors (SSR) on the cell surface, SSR-directed PET/CT is considered the reference standard for imaging of this particular entity. SSR-PET/CT should be the imaging method of choice in every NEN G1 or G2 and considered for re-staging after both potentially curative and non-curative surgeries. The extent of SSR expression is also crucial for determining a patient's eligibility for peptide receptor radionuclide therapy (PRRT). PRRT utilizes [177Lu]Lu-DOTA-TATE to target the SSR receptor and can significantly prolong progression-free survival in patients with advanced, progressive neuroendocrine tumor of the gastroenteropancreatic system (GEP-NET). PET/CT is a central component of the multidisciplinary management of NEN. Variable follow-up intervals are recommended, considering that tumors with higher proliferation rates or advanced metastatic disease require more frequent assessments. The combination with other imaging modalities, like MRI, complements SSR-PET/CT, further enhancing overall diagnostic accuracy. KEY POINTS: Somatostatin receptor-PET/CT (SSR-PET/CT) is the guideline-recommended reference standard for imaging well-differentiated neuroendocrine tumors (NET). SSR-PET/CT should be the diagnostic imaging of choice for staging and post-therapy re-staging of grade 1 or 2 NET (G1 or G2). Variable follow-up intervals are recommended for NET G1 and G2. Tumors with higher proliferation rates or advanced metastatic disease necessitate more frequent assessments.
Collapse
Affiliation(s)
- Ricarda Ebner
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Gabriel T Sheikh
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Mallak N, Yilmaz B, Meyer C, Winters C, Mench A, Jha AK, Prasad V, Mittra E. Theranostics in Neuroendocrine Tumors: Updates and Emerging Technologies. Curr Probl Cancer 2024; 52:101129. [PMID: 39232443 DOI: 10.1016/j.currproblcancer.2024.101129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/22/2024] [Indexed: 09/06/2024]
Abstract
Advancements in somatostatin receptor (SSTR) targeted imaging and treatment of well-differentiated neuroendocrine tumors (NETs) have revolutionized the management of these tumors. This comprehensive review delves into the current practice, discussing the use of the various FDA-approved SSTR-agonist PET tracers and the predictive imaging biomarkers, and elaborating on Lu177-DOTATATE peptide receptor radionuclide therapy (PRRT) including the evolving areas of post-therapy imaging practices, PRRT retreatment, and the potential role of dosimetry in optimizing patient treatments. The future directions sections highlight ongoing research on investigational PET imaging radiotracers, future prospects in alpha particle therapy, and combination therapy strategies.
Collapse
Affiliation(s)
- Nadine Mallak
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Burcak Yilmaz
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Catherine Meyer
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Celeste Winters
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Anna Mench
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Abhinav K Jha
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA; Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Vikas Prasad
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Erik Mittra
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA.
| |
Collapse
|
5
|
Gålne A, Sundlöv A, Enqvist O, Sjögreen Gleisner K, Larsson E, Trägårdh E. Retrospective evaluation of the predictive value of tumour burden at baseline [ 68 Ga]Ga-DOTA-TOC or -TATE PET/CT and tumour dosimetry in GEP-NET patients treated with PRRT. EJNMMI REPORTS 2024; 8:24. [PMID: 39112915 PMCID: PMC11306659 DOI: 10.1186/s41824-024-00210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/17/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE There is a lack of validated imaging biomarkers for prediction of response to peptide receptor radionuclide therapy (PRRT). The primary objective was to evaluate if tumour burden at baseline PET/CT could predict treatment outcomes to PRRT with [177Lu]Lu-DOTA-TATE. Secondary objectives were to evaluate if there was a correlation between tumour burden and mean tumour absorbed dose (AD) during first cycle, and if mean tumour AD or the relative change of tumour burden at first follow-up PET/CT could predict progression free survival (PFS) or overall survival (OS). METHODS Patients with gastroenteropancreatic neuroendocrine tumour (GEP-NET) treated with [177Lu]Lu-DOTA-TATE PRRT were retrospectively included. Tumour burden was quantified from [68 Ga]Ga-DOTA-TOC/TATE PET/CT-images at baseline and first follow-up and expressed as; whole-body somatostatin receptor expressing tumour volume (SRETVwb), total lesion somatostatin receptor expression (TLSREwb), largest tumour lesion diameter and highest SUVmax. The relative change of tumour burden was evaluated in three categories. Mean tumour AD was estimated from the first cycle of PRRT. PFS was defined as time from start of PRRT to radiological or clinical progression. OS was evaluated as time to death. Kaplan Meier survival curves and log-rank test were used to compare PFS and OS between different groups. RESULTS Thirty-one patients had a baseline PET/CT < 6 months before treatment and 25 had a follow-up examination. Median tumour burden was 132 ml (IQR 61-302) at baseline and 71 ml (IQR 36-278) at follow-up. Twenty-two patients had disease progression (median time to progression 17.2 months) and 9 patients had no disease progression (median follow-up 28.7 months). SRETVwb dichotomized by the median at baseline was not associated with longer PFS (p = 0.861) or OS (p = 0.937). Neither TLSREwb, largest tumour lesion or SUVmax showed significant predictive value. There was a moderately strong correlation, however, between SUVmax and mean tumour AD r = 0.705, p < 0.001, but no significant correlation between SRETVwb nor TLSREwb and mean tumour AD. An increase of SRETVwb, TLSREwb or largest tumour lesion at first follow-up PET/CT was significantly correlated with shorter PFS/OS. CONCLUSION Tumour burden at baseline showed no predictive value of PFS/OS after PRRT in this small retrospective study. An increase of tumour burden was predictive of worse outcome.
Collapse
Affiliation(s)
- Anni Gålne
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund and Malmö, Sweden.
- Department of Translational Medicine, Lund University, Malmö, Sweden.
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden.
| | - Anna Sundlöv
- Department of Clinical Sciences, Oncology and Pathology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Olof Enqvist
- Eigenvision AB, Malmö, Sweden
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Erik Larsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Elin Trägårdh
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund and Malmö, Sweden
- Department of Translational Medicine, Lund University, Malmö, Sweden
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden
| |
Collapse
|
6
|
K Nazar A, Basu S. Radiolabeled Somatostatin Analogs for Cancer Imaging. Semin Nucl Med 2024:S0001-2998(24)00058-8. [PMID: 39122608 DOI: 10.1053/j.semnuclmed.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024]
Abstract
Somatostatin receptors (SSTR) are expressed by many tumours especially those related to neuro-endocrine origin and molecular functional imaging of SSTR expression using radiolabelled somatostatin analogs have revolutionized imaging of patients with these group of malignancies. Coming a long way from the first radiolabelled somatostatin analog 123I-Tyr-3-octreotide, there has been significant developments in terms of radionuclides used, the ligands and somatostatin derivatives. 111In-Pentetreotide extensively employed for imaging NETs at the beginning has now been replaced by 68Ga-SSA based PET-CT. SSA-PET/CT performs superior to conventional imaging modalities and has evolved in the mainframe for NET imaging. The advantages were multiple: (i) superior spatial resolution of PET versus SPECT, (ii) quantitative capabilities of PET aiding in disease activity and treatment response monitoring with better precision, (iii) shorter scan time and (iv) less patient exposure to radiation. The modality is indicated for staging, detecting the primary in CUP-NETs, restaging, treatment planning (along with FDG: the concept of dual-tracer PET-CT) as well as treatment response evaluation and follow-up of NETs. SSA PET/CT has also been incorporated in the guidelines for imaging of Pheochromocytoma-Paraganglioma, Medullary carcinoma thyroid, Meningioma and Tumor induced osteomalacia. At present, there is rising interest on (a) 18F-labelled SSA, (b) 64Cu-labelled SSA, and (c) somatostatin antagonists. 18F offers excellent imaging properties, 64Cu makes delayed imaging feasible which has implications in dosimetry and SSTR antagonists bind with the SST receptors with high affinity and specificity, providing high contrast images with less background, which can be translated to theranostics effectively. SSTR have been demonstrated in non-neuroendocrine tumours as well in the peer-reviewed literature, with studies demonstrating the potential of SSA PET/CT in Neuroblastoma, Nasopharyngeal carcinoma, carcinoma prostate (neuroendocrine differentiation) and lymphoma. This review will focus on the currently available SSAs and their history, different SPECT/PET agents, SSTR antagonists, comparison between the various imaging tracers, and their utility in both neuroendocrine and non-neuroendocrine tumors.
Collapse
Affiliation(s)
- Aamir K Nazar
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai.
| |
Collapse
|
7
|
Kaletsch M, Pfestroff A, Luster M. [Nuclear medicine approaches in the diagnosis and treatment of neuroendocrine neoplasms]. Dtsch Med Wochenschr 2024; 149:871-878. [PMID: 39013407 DOI: 10.1055/a-2145-1379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Despite, or perhaps because of the rarity of neuroendocrine neoplasms, the diagnosis and treatment of these malignancies is of particular importance. Nuclear medicine can make an important contribution to this challenge. It offers the most sensitive and specific imaging of these tumor entities and can be helpful in treatment due to the radiotherapeutic drugs that have recently been approved. This theragnostic (fusion of therapeutic and diagnostic) concept is based on the frequent overexpression of somatostatin receptors on neuroendocrine tumor cells.Using diagnostic and therapeutic pharmaceuticals based on analogues from somatostatin, most applications from the nuclear medicine are successful, an additional therapeutic method is SIRT, also known as TARE, in which the hypervascularization of NEN-metastases is used as a therapeutic target.
Collapse
|
8
|
van de Weijer T, Bemer F, de Vos-Geelen J, Hermans B, Mitea C, van der Pol JAJ, Lodewick T, Wildberger JE, Mottaghy FM. Altered biodistribution of [ 68Ga]Ga-DOTA-TOC during somatostatin analogue treatment. Eur J Nucl Med Mol Imaging 2024; 51:2420-2427. [PMID: 38403723 PMCID: PMC11178651 DOI: 10.1007/s00259-024-06659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
PURPOSE The need for an interval between the administration of long-acting Somatostatin Receptor Analogues (SSA) and the [68Ga]Ga-DOTA-TATE PET has been questioned based on recent literature in the new EANM guidelines. Here an earlier studies showed that SSA injection immediately before SSTR PET had minimal effect on normal organ and tumor uptake (1). However, data are scarce and there are (small) differences between [68Ga]Ga-DOTA-TATE and [68Ga]Ga-DOTA-TOC binding affinity, and it remains unknown whether these findings can be directly translated to scans with [68Ga]Ga-DOTA-TOC as well. The purpose of this study was to assess the effect of SSA use on the biodistribution in a subsequent [68Ga]Ga-DOTA-TOC PET/CT and compare this intra-individually across several cycles of SSA treatments. METHODS Retrospectively, 35 patients with NENs were included. [68Ga]Ga-DOTA-TOC PET at staging and after the 1st and 2nd cycle of SSA were included. SUVmean and SUVmax of blood, visceral organs, primary tumor and two metastases were determined. Also, the interval between SSA therapy and the PET scan was registered. RESULTS Treatment with SSA resulted in a significantly higher bloodpool activity and lower visceral tracer uptake. This effect was maintained after a 2nd cycle of SSA therapy. Furthermore, there was an inverse relationship between bloodpool tracer availability and visceral tracer binding and a positive correlation between bloodpool tracer availability and primary tumor tracer uptake. With an interval of up to 5 days, there was a significantly higher bloodpool activity than at longer intervals. CONCLUSION Absolute comparison of the SUV on [68Ga]Ga-DOTA-TOC PET should be done with caution as the altered biodistribution of the tracer after SSA treatment should be taken into account. We recommend not to perform a scan within the first 5 days after the injection of lanreotide.
Collapse
Affiliation(s)
- T van de Weijer
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), University of Maastricht (UM), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - F Bemer
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - J de Vos-Geelen
- Department of Medical Oncology, ENETS Center of Excellence, MUMC+, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School for Oncology and Reproduction (GROW), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - B Hermans
- Department of Medical Oncology, ENETS Center of Excellence, MUMC+, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School for Oncology and Reproduction (GROW), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - C Mitea
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School for Oncology and Reproduction (GROW), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - J A J van der Pol
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - T Lodewick
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - J E Wildberger
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands
| | - F M Mottaghy
- Department of Radiology and Nuclear Medicine, ENETS Center of Excellence, Maastricht University Medical Center (MUMC+), P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands.
- School for Oncology and Reproduction (GROW), UM, P. Debeylaan 25, P.O. Box 5800, 6202, 6229 HX, AZ, Maastricht, The Netherlands.
- Department of Nuclear Medicine, University Hospital RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
9
|
Ebner R, Rübenthaler J, Ricke J, Sheikh GT, Unterrainer LM, Auernhammer CJ, Spitzweg C, Brendel M, Schmid-Tannwald C, Cyran CC. [Imaging of neuroendocrine tumors of the gastrointestinal tract : Value of (hybrid) imaging diagnostics in radiology]. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:553-558. [PMID: 38713221 DOI: 10.1007/s00117-024-01296-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 05/08/2024]
Abstract
CLINICAL/METHODICAL ISSUE Neuroendocrine tumors (NET) represent a heterogeneous group of rare tumors that predominantly arise in the gastrointestinal tract. At the time of initial diagnosis, the NET has already spread locoregionally in about half of the patients, and 27% of patients have already developed distant metastases. Since this plays a crucial role in therapy planning, accurate diagnostic imaging is important. STANDARD RADIOLOGICAL METHODS Due to its high temporal and spatial resolution (multiphasic including arterial phase), computed tomography (CT) plays a decisive role in primary staging and follow-up care, while magnetic resonance imaging (MRI) with its excellent soft tissue contrast offers advantages in the assessment of parenchymal organs in the upper abdomen. METHODICAL INNOVATIONS Somatostatin receptor (SSR) positron emission tomography (PET) provides additional functional information that not only helps to detect the primary tumor and distant metastases, but also has a significant influence on therapeutic management in a theranostic approach. PERFORMANCE Hybrid imaging using SSR-PET/CT has proven to be particularly effective in the detection of NET. Compared to conventional imaging, it provides additional information in 68% of patients, which has a significant impact on clinical management. ACHIEVEMENTS Imaging of NET requires the combined use of various methods such as ultrasound, CT, MRI, and PET/CT to enable accurate diagnosis and effective treatment planning. PRACTICAL RECOMMENDATIONS SSR-PET/CT is a valuable tool for the accurate staging of neuroendocrine tumors of the gastrointestinal tract, especially with small metastases, while MRI with hepatocyte-specific contrast agent and diffusion-weighted imaging is useful for the specific assessment of liver metastases.
Collapse
Affiliation(s)
- R Ebner
- Klinik und Poliklinik für Radiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland.
| | - J Rübenthaler
- Klinik und Poliklinik für Radiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland
- Interdisziplinäres Zentrum für Neuroendokrine Tumoren des Gastroenteropankreatischen Systems (GEPNET-KUM), LMU Klinikum, LMU München, München, Deutschland
| | - J Ricke
- Klinik und Poliklinik für Radiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland
- Interdisziplinäres Zentrum für Neuroendokrine Tumoren des Gastroenteropankreatischen Systems (GEPNET-KUM), LMU Klinikum, LMU München, München, Deutschland
| | - G T Sheikh
- Klinik und Poliklinik für Nuklearmedizin, LMU Klinikum, LMU München, München, Deutschland
| | - L M Unterrainer
- Klinik und Poliklinik für Nuklearmedizin, LMU Klinikum, LMU München, München, Deutschland
| | - C J Auernhammer
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, München, Deutschland
- Interdisziplinäres Zentrum für Neuroendokrine Tumoren des Gastroenteropankreatischen Systems (GEPNET-KUM), LMU Klinikum, LMU München, München, Deutschland
| | - C Spitzweg
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, München, Deutschland
- Interdisziplinäres Zentrum für Neuroendokrine Tumoren des Gastroenteropankreatischen Systems (GEPNET-KUM), LMU Klinikum, LMU München, München, Deutschland
| | - M Brendel
- Klinik und Poliklinik für Nuklearmedizin, LMU Klinikum, LMU München, München, Deutschland
| | - C Schmid-Tannwald
- Klinik und Poliklinik für Radiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland
| | - C C Cyran
- Klinik und Poliklinik für Radiologie, LMU Klinikum, LMU München, Marchioninistr. 15, 81377, München, Deutschland
- Interdisziplinäres Zentrum für Neuroendokrine Tumoren des Gastroenteropankreatischen Systems (GEPNET-KUM), LMU Klinikum, LMU München, München, Deutschland
| |
Collapse
|
10
|
Sakellis C, Jacene HA. Neuroendocrine Tumors: Diagnostics. PET Clin 2024; 19:325-339. [PMID: 38714399 DOI: 10.1016/j.cpet.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024]
Abstract
Neuroendocrine neoplasms (NEN) are rare tumors arising from neuroendocrine cells. NEN are ideally suited for a theragnostic approach due to their specific expression of somatostatin receptors (SSTR). SSTR imaging of NEN dates back to the 1980s, but has evolved recently due to the introduction of more sensitive SSTR PET radiotracers. SSTR PET is a primary imaging modality for identifying NEN and characterizing SSTR expression. SSTR PET is complementary to anatomic imaging for assessing tumor response to treatment. SSTR PET is mandated to determine eligibility for peptide receptor radionuclide therapy. Here, the role of imaging to aid management of NEN is reviewed.
Collapse
Affiliation(s)
- Christopher Sakellis
- Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Avenue, DL198, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02215, USA
| | - Heather A Jacene
- Department of Imaging, Dana-Farber Cancer Institute, 450 Brookline Avenue, DL198, Boston, MA 02215, USA; Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Firsova M, Treglia G, Sempoux C, Dromain C, Prior JO, Schaefer N, Boughdad S. Increased [ 68Ga]Ga-SST uptake in the uncinate pancreatic process in new digital PET/CT machine and potential association with clinical and histologic factors in NET patients. EJNMMI REPORTS 2024; 8:18. [PMID: 38910232 PMCID: PMC11194230 DOI: 10.1186/s41824-024-00203-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION A physiological increase in the uptake of [68Ga]Ga-labeled somatostatin analogues ([68Ga]Ga-SST) PET tracers has been reported in the uncinate pancreatic process (UP) and might be even higher in latest generation of PET/CT scanners and might be falsely interpreted as NET. We aimed to investigate the uptake of UP in a large population of NET patients who underwent [68Ga]Ga-SST PET/CT with digital SiPM detectors. We also explored potential associations between UP uptake and various clinical, imaging, and pathological factors routinely assessed in NET patients. METHODS We analyzed all consecutive NET patients from July 2018 to June 2022 in this retrospective, single-center study. All patients underwent a [68Ga]Ga-SST PET/CT scan on a digital SiPM PET/CT scanner. On visual analysis, we distinguished between normal linear and homogenous UP uptake or abnormal if otherwise. We compared SUVmax/mean in patients with normal UP uptake to those with abnormal UP uptake with suspicious NET lesions on contrast-enhanced CT (ce-CT) and according to the site of the primary NET (pancreatic NET vs. other), patient gender (female vs. male) and tumor grade (grade 1-2 vs. 3) using a Mann-Whitney test. We also assessed the correlation between SUVmax/mean values in UP with patients' age, primary NET Ki-67 counting, and its SUVmax/mean, TLA and MTV values. RESULTS We included 131 NET patients with a total of 34 [68Ga]Ga-DOTATATE PET/CT and 113 [68Ga]Ga-DOTATOC PET/CT scans. An abnormal UP uptake was seen in 32 patients with 65.7% of suspicious NET lesion or extrinsic compression on morphological imaging. Normal UP uptake SUVmax/mean were measured in 115 [68Ga]Ga-SST scans (78.2%) with normal UP uptake and without suspicious lesion on morphological imaging. We found an average SUVmax of 12.3 ± 4.1 for [68Ga]Ga-DOTATATE and 19.8 ± 9.8 g/ml for [68Ga]Ga-DOTATOC, hence higher than those reported in the literature [SUVmax 5 ± 1.6 to 12.6 ± 2.2 g/ml] with significant difference with abnormal UP uptake and between both PET tracers (both p < 0.01). Significant results were a higher UP uptake on [68Ga]Ga-DOTATOC in male patients (p = 0.02) and significant associations between UP uptake on [68Ga]Ga-DOTATOC and SUVmax/mean of the primary tumor (ρ [0.337-0.363]; p [0.01-0.02]). CONCLUSION We confirmed a higher and very frequent UP uptake in latest SiPM-detector [68Ga]Ga-SST PET/CT with an even higher uptake in patients that had [68Ga]Ga-DOTATOC PET/CT. SUVmean/max were significantly higher in abnormal UP uptake but there were overlaps with UP SUV values for both [68Ga]Ga-SST and a correlation to morphological imaging is crucial. Besides, significant associations between UP uptake and SUVmean/max of the primary NET as well as patients' gender were seen in the larger cohort of [68Ga]Ga-DOTATOC patients suggesting that both physiological and pathological parameters could affect UP uptake.
Collapse
Affiliation(s)
- Maria Firsova
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Giorgio Treglia
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Via A. Gallino 12, 6500, Bellinzona, Switzerland
- Academic Education, Research and Innovation Area, General Directorate, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Christine Sempoux
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Clarisse Dromain
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sarah Boughdad
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| |
Collapse
|
12
|
Sharma A, Singh LR. An insight into the pharmacology of cysteine/methionine containing peptide drugs. Eur J Med Chem 2024; 271:116456. [PMID: 38691890 DOI: 10.1016/j.ejmech.2024.116456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Since last century, peptides have emerged as potential drugs with >90 FDA approvals for various targets with several in the pipeline. Sulphur, in peptides is present either as thiol (-SH) from Cys or thioether from Met. In this review, all the peptides approved by FDA since 2000 containing sulphur have been included. Among them ∼50 % contains disulphide bridges. This clearly demonstrates the significance of disulphide bonds in peptide drugs. This can be achieved synthetically by using orthogonal protecting groups (PGs) for -SH. These PGs are compatible with Solid Phase Peptide Synthesis (SPPS), which is still the method of choice for peptide synthesis. The orthogonal PGs used for Cys thiol side chain protecting for disulphide bond formation have been included which are currently in use both by academia and industry from small scale to large scale synthesis. In addition, the details of the FDA approved drugs containing Cys and Met (or both) have also been discussed.
Collapse
Affiliation(s)
- Anamika Sharma
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India; Chemical Science Division, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - L Ravithej Singh
- Chemical Science Division, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Fluoro-Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| |
Collapse
|
13
|
Mallak N, O'Brien SR, Pryma DA, Mittra E. Theranostics in Neuroendocrine Tumors. Cancer J 2024; 30:185-193. [PMID: 38753753 DOI: 10.1097/ppo.0000000000000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Neuroendocrine tumors (NETs) are rare tumors that develop from cells of the neuroendocrine system and can originate in multiple organs and tissues such as the bowels, pancreas, adrenal glands, ganglia, thyroid, and lungs. This review will focus on gastroenteropancreatic NETs (more commonly called NETs) characterized by frequent somatostatin receptor (SSTR) overexpression and pheochromocytomas/paragangliomas (PPGLs), which typically overexpress norepinephrine transporter. Advancements in SSTR-targeted imaging and treatment have revolutionized the management of patients with NETs. This comprehensive review delves into the current practice, discussing the use of the various Food and Drug Administration-approved SSTR-agonist positron emission tomography tracers and the predictive imaging biomarkers, and elaborating on 177Lu-DOTATATE peptide receptor radionuclide therapy including the evolving areas of posttherapy imaging practices and peptide receptor radionuclide therapy retreatment. SSTR-targeted imaging and therapy can also be used in patients with PPGL; however, this patient population has demonstrated the best outcomes from norepinephrine transporter-targeted therapy with 131I-metaiodobenzylguanidine. Metaiodobenzylguanidine theranostics for PPGL will be discussed, noting that in 2024 it became commercially unavailable in the United States. Therefore, the use and reported success of SSTR theranostics for PPGL will also be explored.
Collapse
Affiliation(s)
- Nadine Mallak
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| | - Sophia R O'Brien
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Daniel A Pryma
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Erik Mittra
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| |
Collapse
|
14
|
Müller PC, Pfister M, Eshmuminov D, Lehmann K. Liver transplantation as an alternative for the treatment of neuroendocrine liver metastasis: Appraisal of the current evidence. Hepatobiliary Pancreat Dis Int 2024; 23:146-153. [PMID: 37634987 DOI: 10.1016/j.hbpd.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/10/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Liver transplantation (LT) for neuroendocrine liver metastases (NELM) is still in debate. Studies comparing LT with liver resection (LR) for NELM are scarce, as patient selection is heterogeneous and experience is limited. The goal of this review was to provide a critical analysis of the evidence on LT versus LR in the treatment of NELM. DATA SOURCES A scoping literature search on LT and LR for NELM was performed with PubMed, including English articles up to March 2023. RESULTS International guidelines recommend LR for NELM in resectable, well-differentiated tumors in the absence of extrahepatic metastatic disease with superior results of LR compared to systemic or liver-directed therapies. Advanced liver surgery has extended resectability criteria whilst entailing increased perioperative risk and short disease-free survival. In highly selected patients (based on the Milan criteria) with unresectable NELM, oncologic results of LT are promising. Prognostic factors include tumor biology (G1/G2) and burden, waiting time for LT, patient age and extrahepatic spread. Based on low-level evidence, LT for low-grade NELM within the Milan criteria resulted in improved disease-free survival and overall survival compared to LR. The benefits of LT were lost in patients beyond the Milan NELM-criteria. CONCLUSIONS With adherence to strict selection criteria especially tumor biology, LT for NELM is becoming a valuable option providing oncologic benefits compared to LR. Recent evidence suggests even stricter selection criteria with regard to tumor biology.
Collapse
Affiliation(s)
- Philip C Müller
- Department of Surgery and Transplantation, University Hospital Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Matthias Pfister
- Department of Surgery and Transplantation, University Hospital Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Dilmurodjon Eshmuminov
- Department of Surgery and Transplantation, University Hospital Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland
| | - Kuno Lehmann
- Department of Surgery and Transplantation, University Hospital Zurich, Rämistrasse 100, Zurich CH-8091, Switzerland.
| |
Collapse
|
15
|
Harini K, Jayanthi MR, Hari KSV, Batchu S. A Rare Presentation of Extrahepatic Biliary Neuroendocrine Tumor Diagnosed using 68Ga-DOTA-TOC Imaging, But Undetectable on 68Ga-FAPI Imaging. Indian J Nucl Med 2024; 39:155-157. [PMID: 38989315 PMCID: PMC11232718 DOI: 10.4103/ijnm.ijnm_149_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 07/12/2024] Open
Abstract
Neuroendocrine tumors (NETs) are commonly seen in the small intestine and rarely found within the bile ducts. This low incidence is due to a smaller number of Kulchitsky cells in the extrahepatic biliary tree, which predisposes to the disease. The diagnosis of biliary tree carcinoid preoperatively is very rare, with most cases in the literature being incidentally diagnosed during surgery or being identified on the histopathology report postoperatively. Here, we present an interesting case of an extrahepatic biliary NET which was diagnosed preoperatively.
Collapse
Affiliation(s)
- Koorma Harini
- Department of Nuclear Medicine and PET-CT, AIG Hospitals, Hyderabad, Telangana, India
| | - Mohan Roop Jayanthi
- Department of Nuclear Medicine and PET-CT, AIG Hospitals, Hyderabad, Telangana, India
| | - K S Vishnu Hari
- Department of Nuclear Medicine and PET-CT, AIG Hospitals, Hyderabad, Telangana, India
| | - Suneetha Batchu
- Department of Nuclear Medicine and PET-CT, AIG Hospitals, Hyderabad, Telangana, India
| |
Collapse
|
16
|
Dubash S, Barwick TD, Kozlowski K, Rockall AG, Khan S, Khan S, Yusuf S, Lamarca A, Valle JW, Hubner RA, McNamara MG, Frilling A, Tan T, Wernig F, Todd J, Meeran K, Pratap B, Azeem S, Huiban M, Keat N, Lozano-Kuehne JP, Aboagye EO, Sharma R. Somatostatin Receptor Imaging with [ 18F]FET-βAG-TOCA PET/CT and [ 68Ga]Ga-DOTA-Peptide PET/CT in Patients with Neuroendocrine Tumors: A Prospective, Phase 2 Comparative Study. J Nucl Med 2024; 65:jnumed.123.266601. [PMID: 38331457 PMCID: PMC10924162 DOI: 10.2967/jnumed.123.266601] [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: 09/22/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 02/10/2024] Open
Abstract
There is a clinical need for 18F-labeled somatostatin analogs for the imaging of neuroendocrine tumors (NET), given the limitations of using [68Ga]Ga-DOTA-peptides, particularly with regard to widespread accessibility. We have shown that [18F]fluoroethyl-triazole-[Tyr3]-octreotate ([18F]FET-βAG-TOCA) has favorable dosimetry and biodistribution. As a step toward clinical implementation, we conducted a prospective, noninferiority study of [18F]FET-βAG-TOCA PET/CT compared with [68Ga]Ga-DOTA- peptide PET/CT in patients with NET. Methods: Forty-five patients with histologically confirmed NET, grades 1 and 2, underwent PET/CT imaging with both [18F]FET-βAG-TOCA and [68Ga]Ga-peptide performed within a 6-mo window (median, 77 d; range, 6-180 d). Whole-body PET/CT was conducted 50 min after injection of 165 MBq of [18F]FET-βAG-TOCA. Tracer uptake was evaluated by comparing SUVmax and tumor-to-background ratios at both lesion and regional levels by 2 unblinded, experienced readers. A randomized, blinded reading of both scans was also then undertaken by 3 experienced readers, and consensus was assessed at a regional level. The ability of both tracers to visualize liver metastases was also assessed. Results: A total of 285 lesions were detected on both imaging modalities. An additional 13 tumor deposits were seen in 8 patients on [18F]FET-βAG-TOCA PET/CT, and [68Ga]Ga-DOTA-peptide PET/CT detected an additional 7 lesions in 5 patients. Excellent correlation in SUVmax was observed between both tracers (r = 0.91; P < 0.001). No difference was observed between median SUVmax across regions, except in the liver, where the median tumor-to-background ratio of [18F]FET-βAG-TOCA was significantly lower than that of [68Ga]Ga-DOTA-peptide (2.5 ± 1.9 vs. 3.5 ± 2.3; P < 0.001). Conclusion: [18F]FET-βAG-TOCA was not inferior to [68Ga]Ga-DOTA-peptide in visualizing NET and may be considered in routine clinical practice given the longer half-life and availability of the cyclotron-produced fluorine radioisotope.
Collapse
Affiliation(s)
- Suraiya Dubash
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Tara D Barwick
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Imaging, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Kasia Kozlowski
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Andrea G Rockall
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Sairah Khan
- Department of Imaging, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Sameer Khan
- Department of Imaging, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Siraj Yusuf
- Radiology and Nuclear Medicine Department, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Angela Lamarca
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Richard A Hubner
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mairéad G McNamara
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Andrea Frilling
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Tricia Tan
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Florian Wernig
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jeannie Todd
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Karim Meeran
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Bhavesh Pratap
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Saleem Azeem
- Invicro-London, Imperial College London, London, United Kingdom; and
| | - Michael Huiban
- Invicro-London, Imperial College London, London, United Kingdom; and
| | - Nicholas Keat
- Invicro-London, Imperial College London, London, United Kingdom; and
| | - Jingky P Lozano-Kuehne
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Population Health Sciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle, United Kingdom
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Rohini Sharma
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom;
| |
Collapse
|
17
|
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: 4.0] [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
|
18
|
Mourtada F, Tomiyoshi K, Sims-Mourtada J, Mukai-Sasaki Y, Yagihashi T, Namiki Y, Murai T, Yang DJ, Inoue T. Actinium-225 Targeted Agents: Where Are We Now? Brachytherapy 2023; 22:697-708. [PMID: 37690972 PMCID: PMC10840862 DOI: 10.1016/j.brachy.2023.06.228] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 09/12/2023]
Abstract
α-particle targeted radionuclide therapy has shown promise for optimal cancer management, an exciting new era for brachytherapy. Alpha-emitting nuclides can have significant advantages over gamma- and beta-emitters due to their high linear energy transfer (LET). While their limited path length results in more specific tumor 0kill with less damage to surrounding normal tissues, their high LET can produce substantially more lethal double strand DNA breaks per radiation track than beta particles. Over the last decade, the physical and chemical attributes of Actinium-225 (225Ac) including its half-life, decay schemes, path length, and straightforward chelation ability has peaked interest for brachytherapy agent development. However, this has been met with challenges including source availability, accurate modeling for standardized dosimetry for brachytherapy treatment planning, and laboratory space allocation in the hospital setting for on-demand radiopharmaceuticals production. Current evidence suggests that a simple empirical approach based on 225Ac administered radioactivity may lead to inconsistent outcomes and toxicity. In this review article, we highlight the recent advances in 225Ac source production, dosimetry modeling, and current clinical studies.
Collapse
Affiliation(s)
- Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA.
| | - Katsumi Tomiyoshi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | | | - Yuki Mukai-Sasaki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Takayuki Yagihashi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Taro Murai
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - David J Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| |
Collapse
|
19
|
Halfdanarson TR, Mallak N, Paulson S, Chandrasekharan C, Natwa M, Kendi AT, Kennecke HF. Monitoring and Surveillance of Patients with Gastroenteropancreatic Neuroendocrine Tumors Undergoing Radioligand Therapy. Cancers (Basel) 2023; 15:4836. [PMID: 37835530 PMCID: PMC10571645 DOI: 10.3390/cancers15194836] [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: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Radioligand therapy (RLT) with [177Lu]Lu-DOTA-TATE is a standard of care for adult patients with somatostatin-receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Taking advantage of this precision nuclear medicine approach requires diligent monitoring and surveillance, from the use of diagnostic SSTR-targeted radioligand imaging for the selection of patients through treatment and assessments of response. Published evidence-based guidelines assist the multidisciplinary healthcare team by providing acceptable approaches to care; however, the sheer heterogeneity of GEP-NETs can make these frameworks difficult to apply in individual clinical circumstances. There are also contradictions in the literature regarding the utility of novel approaches in monitoring and surveilling patients with GEP-NETs receiving RLT. This article discusses the emerging evidence on imaging, clinical biochemistry, and tumor assessment criteria in the management of patients receiving RLT for GEP-NETs; additionally, it documents our own best practices. This allows us to offer practical guidance on how to effectively implement monitoring and surveillance measures to aid patient-tailored clinical decision-making.
Collapse
Affiliation(s)
| | - Nadine Mallak
- Division of Molecular Imaging and Therapy, Oregon Health and Science University, Portland, OR 97239, USA;
| | | | | | - Mona Natwa
- Langone Health, New York University, New York, NY 10016, USA
| | | | | |
Collapse
|
20
|
Fabritius MP, Soltani V, Cyran CC, Ricke J, Bartenstein P, Auernhammer CJ, Spitzweg C, Schnitzer ML, Ebner R, Mansournia S, Hinterberger A, Lohse A, Sheikh GT, Winkelmann M, Knösel T, Ingenerf M, Schmid-Tannwald C, Kunz WG, Rübenthaler J, Grawe F. Diagnostic accuracy of SSR-PET/CT compared to histopathology in the identification of liver metastases from well-differentiated neuroendocrine tumors. Cancer Imaging 2023; 23:92. [PMID: 37770958 PMCID: PMC10537814 DOI: 10.1186/s40644-023-00614-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Histopathology is the reference standard for diagnosing liver metastases of neuroendocrine tumors (NETs). Somatostatin receptor-positron emission tomography / computed tomography (SSR-PET/CT) has emerged as a promising non-invasive imaging modality for staging NETs. We aimed to assess the diagnostic accuracy of SSR-PET/CT in the identification of liver metastases in patients with proven NETs compared to histopathology. METHODS Histopathologic reports of 139 resected or biopsied liver lesions of patients with known NET were correlated with matching SSR-PET/CTs and the positive/negative predictive value (PPV/NPV), sensitivity, specificity, and diagnostic accuracy of SSR-PET/CT were evaluated. PET/CT reading was performed by one expert reader blinded to histopathology and clinical data. RESULTS 133 of 139 (95.7%) liver lesions showed malignant SSR-uptake in PET/CT while initial histopathology reported on 'liver metastases of NET´ in 127 (91.4%) cases, giving a PPV of 91.0%. Re-biopsy of the initially histopathologically negative lesions (reference standard) nevertheless diagnosed 'liver metastases of NET' in 6 cases, improving the PPV of PET/CT to 95.5%. Reasons for initial false-negative histopathology were inadequate sampling in the sense of non-target biopsies. The 6 (4.3%) SSR-negative lesions were all G2 NETs with a Ki-67 between 2-15%. CONCLUSION SSR-PET/CT is a highly accurate imaging modality for the diagnosis of liver metastases in patients with proven NETs. However, we found that due to the well-known tumor heterogeneity of NETs, specifically in G2 NETs approximately 4-5% are SSR-negative and may require additional imaging with [18F]FDG PET/CT.
Collapse
Affiliation(s)
- M P Fabritius
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - V Soltani
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - C C Cyran
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - J Ricke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - C J Auernhammer
- Department of Internal Medicine 4, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - C Spitzweg
- Department of Internal Medicine 4, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - M L Schnitzer
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - R Ebner
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - S Mansournia
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - A Hinterberger
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - A Lohse
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - G T Sheikh
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
| | - M Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - T Knösel
- Department of Pathology, University Hospital, LMU Munich, 81377, Munich, Germany
| | - M Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - C Schmid-Tannwald
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - W G Kunz
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - J Rübenthaler
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Freba Grawe
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany.
| |
Collapse
|
21
|
Kalaitzidis P, Gustafsson J, Hindorf C, Ljungberg M. Monte Carlo investigation of PET [ 68Ga]Ga-DOTA-TOC activity-administration protocols for consistent image quality. Heliyon 2023; 9:e19504. [PMID: 37681181 PMCID: PMC10480678 DOI: 10.1016/j.heliyon.2023.e19504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
One example of a PET exam that suffers from noise problems is [68Ga]Ga-DOTA-TOC, where patients are generally administered between 100 and 200 MBq [68Ga]Ga-DOTA-TOC, irrespective of size. However, a fixed activity can result in low signal-to-noise ratios (SNRs) in larger patients. This study aimed to evaluate the impact on image quality with respect to injected activity and patient habitus through Monte Carlo (MC) simulation. Eight anthropomorphic computer phantoms with body mass indices (BMIs) between 19 kg/m2 and 38 kg/m2 and tumours distributed in the liver were simulated using the MC software Gate v8.2 with an activity distribution defined according to [68Ga]Ga-DOTA-TOC standardised uptake values. Three activity-administration protocols were simulated: (i) with a fixed activity of 100 MBq, (ii) with the activity scaled by 2 MBq/kg, and (iii) with the activity scaled by a body size-dependent power-function based on the SNR obtained with (ii). BMI, weight, body surface area, and abdominal circumference were evaluated body size parameters. Images were reconstructed with the CASToR software and evaluated for background SNR and lesion contrast-to-noise ratio (CNR). Large SNR variabilities were obtained with protocols (i) and (ii), while (iii) generated good consistency. Several tumours failed to reach a CNR of 5 for large phantoms with protocol (i), but the CNR was generally improved by (ii) and (iii). An activity scaled by patient habitus generate better image quality consistency, which increases the likelihood that patients receive a similar standard of care.
Collapse
Affiliation(s)
| | | | - Cecilia Hindorf
- Medical Radiation Physics, Lund, Lund University, Lund, Sweden
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | |
Collapse
|
22
|
Navin PJ, Ehman EC, Liu JB, Halfdanarson TR, Gupta A, Laghi A, Yoo DC, Carucci LR, Schima W, Sheedy SP. Imaging of Small-Bowel Neuroendocrine Neoplasms: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2023; 221:289-301. [PMID: 36752369 DOI: 10.2214/ajr.22.28877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Neuroendocrine neoplasms (NENs) of the small bowel are typically slow-growing lesions that remain asymptomatic until reaching an advanced stage. Imaging modalities for lesion detection, staging, and follow-up in patients with known or suspected NEN include CT enterography, MR enterography, and PET/CT using a somatostatin receptor analog. FDG PET/CT may have a role in the evaluation of poorly differentiated NENs. Liver MRI, ideally with a hepatocyte-specific contrast agent, should be used in the evaluation of hepatic metastases. Imaging informs decisions regarding both surgical approaches and systematic therapy (specifically, peptide receptor radionuclide therapy). This AJR Expert Panel Narrative Review describes the multimodality imaging features of small-bowel NENs; explores the optimal imaging modalities for their diagnosis, staging, and follow-up; and discusses how imaging may be used to guide therapy.
Collapse
Affiliation(s)
- Patrick J Navin
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Eric C Ehman
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Jason B Liu
- Department of Surgery, Division of Surgical Oncology, Brigham and Women's Hospital, Boston, MA
| | | | - Akshya Gupta
- Department of Imaging Sciences, University of Rochester, Rochester, NY
| | - Andrea Laghi
- Department of Medical Surgical Sciences and Translational Medicine, AOU Sant'Andrea, Sapienza University of Rome, Rome, Italy
| | - Don C Yoo
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Laura R Carucci
- Department of Radiology, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Wolfgang Schima
- Department of Diagnostic and Interventional Radiology, Goettlicher Heiland Krankenhaus, Barmherzige Schwestern Krankenhaus and Sankt Josef Krankenhaus, Vienna, Austria
| | - Shannon P Sheedy
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| |
Collapse
|
23
|
Nhàn NTT, Yamada T, Yamada KH. Peptide-Based Agents for Cancer Treatment: Current Applications and Future Directions. Int J Mol Sci 2023; 24:12931. [PMID: 37629112 PMCID: PMC10454368 DOI: 10.3390/ijms241612931] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Peptide-based strategies have received an enormous amount of attention because of their specificity and applicability. Their specificity and tumor-targeting ability are applied to diagnosis and treatment for cancer patients. In this review, we will summarize recent advancements and future perspectives on peptide-based strategies for cancer treatment. The literature search was conducted to identify relevant articles for peptide-based strategies for cancer treatment. It was performed using PubMed for articles in English until June 2023. Information on clinical trials was also obtained from ClinicalTrial.gov. Given that peptide-based strategies have several advantages such as targeted delivery to the diseased area, personalized designs, relatively small sizes, and simple production process, bioactive peptides having anti-cancer activities (anti-cancer peptides or ACPs) have been tested in pre-clinical settings and clinical trials. The capability of peptides for tumor targeting is essentially useful for peptide-drug conjugates (PDCs), diagnosis, and image-guided surgery. Immunomodulation with peptide vaccines has been extensively tested in clinical trials. Despite such advantages, FDA-approved peptide agents for solid cancer are still limited. This review will provide a detailed overview of current approaches, design strategies, routes of administration, and new technological advancements. We will highlight the success and limitations of peptide-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL 60607, USA
| | - Kaori H. Yamada
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology & Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Gålne A, Enqvist O, Sundlöv A, Valind K, Minarik D, Trägårdh E. AI-based quantification of whole-body tumour burden on somatostatin receptor PET/CT. Eur J Hybrid Imaging 2023; 7:14. [PMID: 37544941 PMCID: PMC10404578 DOI: 10.1186/s41824-023-00172-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Segmenting the whole-body somatostatin receptor-expressing tumour volume (SRETVwb) on positron emission tomography/computed tomography (PET/CT) images is highly time-consuming but has shown value as an independent prognostic factor for survival. An automatic method to measure SRETVwb could improve disease status assessment and provide a tool for prognostication. This study aimed to develop an artificial intelligence (AI)-based method to detect and quantify SRETVwb and total lesion somatostatin receptor expression (TLSREwb) from [68Ga]Ga-DOTA-TOC/TATE PET/CT images. METHODS A UNet3D convolutional neural network (CNN) was used to train an AI model with [68Ga]Ga-DOTA-TOC/TATE PET/CT images, where all tumours were manually segmented with a semi-automatic method. The training set consisted of 148 patients, of which 108 had PET-positive tumours. The test group consisted of 30 patients, of which 25 had PET-positive tumours. Two physicians segmented tumours in the test group for comparison with the AI model. RESULTS There were good correlations between the segmented SRETVwb and TLSREwb by the AI model and the physicians, with Spearman rank correlation coefficients of r = 0.78 and r = 0.73, respectively, for SRETVwb and r = 0.83 and r = 0.81, respectively, for TLSREwb. The sensitivity on a lesion detection level was 80% and 79%, and the positive predictive value was 83% and 84% when comparing the AI model with the two physicians. CONCLUSION It was possible to develop an AI model to segment SRETVwb and TLSREwb with high performance. A fully automated method makes quantification of tumour burden achievable and has the potential to be more widely used when assessing PET/CT images.
Collapse
Affiliation(s)
- Anni Gålne
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden.
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden.
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden.
| | - Olof Enqvist
- Eigenvision AB, Malmö, Sweden
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Anna Sundlöv
- Department of Clinical Sciences, Oncology and Pathology, Lund University, Lund, Sweden
| | - Kristian Valind
- Department of Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden
| | - David Minarik
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden
- Radiation Physics, Skåne University Hospital, Malmö, Sweden
| | - Elin Trägårdh
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
- WCMM Wallenberg Centre for Molecular Medicine, Lund, Sweden
- Department of Medical Imaging and Physiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
25
|
Zhu T, Hsu JC, Guo J, Chen W, Cai W, Wang K. Radionuclide-based theranostics - a promising strategy for lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:2353-2374. [PMID: 36929181 PMCID: PMC10272099 DOI: 10.1007/s00259-023-06174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE This review aims to provide a comprehensive overview of the latest literature on personalized lung cancer management using different ligands and radionuclide-based tumor-targeting agents. BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. Due to the heterogeneity of lung cancer, advances in precision medicine may enhance the disease management landscape. More recently, theranostics using the same molecule labeled with two different radionuclides for imaging and treatment has emerged as a promising strategy for systemic cancer management. In radionuclide-based theranostics, the target, ligand, and radionuclide should all be carefully considered to achieve an accurate diagnosis and optimal therapeutic effects for lung cancer. METHODS We summarize the latest radiotracers and radioligand therapeutic agents used in diagnosing and treating lung cancer. In addition, we discuss the potential clinical applications and limitations associated with target-dependent radiotracers as well as therapeutic radionuclides. Finally, we provide our views on the perspectives for future development in this field. CONCLUSIONS Radionuclide-based theranostics show great potential in tailored medical care. We expect that this review can provide an understanding of the latest advances in radionuclide therapy for lung cancer and promote the application of radioligand theranostics in personalized medicine.
Collapse
Affiliation(s)
- Tianxing Zhu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
26
|
Loken EK, Huang RY. Advanced Meningioma Imaging. Neurosurg Clin N Am 2023; 34:335-345. [PMID: 37210124 DOI: 10.1016/j.nec.2023.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Noninvasive imaging methods are used to accurately diagnose meningiomas and track their growth and location. These techniques, including computed tomography, MRI, and nuclear medicine, are also being used to gather more information about the biology of the tumors and potentially predict their grade and impact on prognosis. In this article, we will discuss the current and developing uses of these imaging techniques including additional analysis using radiomics in the diagnosis and treatment of meningiomas, including treatment planning and prediction of tumor behavior.
Collapse
Affiliation(s)
- Erik K Loken
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
27
|
Milosevic A, Styczen H, Grueneisen J, Li Y, Weber M, Fendler WP, Kirchner J, Damman P, Wrede K, Lazaridis L, Glas M, Guberina M, Eckstein A, Blau T, Herrmann K, Umutlu L, Forsting M, Deuschl C, Schaarschmidt B. Evaluation of [ 68Ga]-DOTATOC PET/MRI in Patients with Meningioma of the Subcranial and Intraorbital Space. J Nucl Med 2023:jnumed.123.265424. [PMID: 37385668 DOI: 10.2967/jnumed.123.265424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/20/2023] [Indexed: 07/01/2023] Open
Abstract
Meningiomas are known to express somatostatin receptor (SSTR) type 2 to a high degree. Therefore, radiolabeled somatostatin analogs, such as DOTATOC, have been introduced for PET imaging of meningiomas. However, the benefit of hybrid SSTR PET/MRI is still debated. Here, we report our experience with [68Ga]-DOTATOC PET/MRI. Methods: PET/MRI was performed in 60 patients with suspected or diagnosed meningiomas of the skull plane and eye socket. Acquired datasets were reported by 2 independent readers regarding local tumor extent and signal characteristics. Histopathologic results and follow-up imaging served as the reference standard. SUVs of target lesions were analyzed according to the corresponding maximal tracer uptake. The diagnostic accuracy of PET/MRI and conventional MRI was determined independently and compared with the reference standard. Results: In total, 60 target lesions were identified, with 54 considered to be meningiomas according to the reference standard. Sensitivity and specificity of PET/MRI versus MRI alone were 95% versus 96% and 75% versus 66%, respectively. The McNemar test was not able to distinguish any differences between PET/MRI and the reference standard or MRI and the reference standard. No differences were found between the 2 modalities with respect to local infiltration. Conclusion: SSTR PET/MRI and MRI yielded similar accuracy for the detection of meningiomas of the skull base and intraorbital space. Here, sequential low-dose SSTR PET/CT might be helpful for the planning of radioligand therapy or radiotherapy.
Collapse
Affiliation(s)
- Aleksandar Milosevic
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany;
| | - Hanna Styczen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Johannes Grueneisen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Yan Li
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Manuel Weber
- Department of Nuclear Medicine, University Hospital Essen, Düsseldorf, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Düsseldorf, Germany
| | - Julian Kirchner
- Institute of Radiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Philipp Damman
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Düsseldorf, Germany
| | - Karsten Wrede
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Düsseldorf, Germany
| | - Lazaros Lazaridis
- Department of Neurology and Neurooncology, University Hospital Essen, Düsseldorf, Germany
| | - Martin Glas
- Department of Neurology and Neurooncology, University Hospital Essen, Düsseldorf, Germany
| | - Maja Guberina
- Department of Radiotherapy, University Hospital Essen, Düsseldorf, Germany
| | - Anja Eckstein
- Department of Ophthalmology, University Hospital Essen, Düsseldorf, Germany; and
| | - Tobias Blau
- Department of Neuropathology, University Hospital Essen, Düsseldorf, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Düsseldorf, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Cornelius Deuschl
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| | - Benedikt Schaarschmidt
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Düsseldorf, Germany
| |
Collapse
|
28
|
Adnan A, Basu S. Somatostatin Receptor Targeted PET-CT and Its Role in the Management and Theranostics of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics (Basel) 2023; 13:2154. [PMID: 37443548 DOI: 10.3390/diagnostics13132154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Somatostatin receptor (SSTR) agonist-based Positron Emission Tomography-Computed Tomography (PET-CT) imaging is nowadays the mainstay for the assessment and diagnostic imaging of neuroendocrine neoplasms (NEN), especially in well-differentiated neuroendocrine tumors (NET) (World Health Organization (WHO) grade I and II). Major clinical indications for SSTR imaging are primary staging and metastatic workup, especially (a) before surgery, (b) detection of unknown primary in metastatic NET, (c) patient selection for theranostics and appropriate therapy, especially peptide receptor radionuclide therapy (PRRT), while less major indications include treatment response evaluation on and disease prognostication. Dual tracer PET-CT imaging using SSTR targeted PET tracers, viz. [68Ga]Ga-DOTA-Tyr3-Octreotate (DOTA-TATE) and [68Ga]Ga-DOTA-NaI3-Octreotide (DOTA-NOC), and fluorodeoxyglucose (FDG), have recently gained widespread acceptance for better assessment of whole-body tumor biology compared to single-site histopathology, in terms of being non-invasive and the ability to assess inter- and intra-tumoral heterogeneity on a global scale. FDG uptake has been identified as independent adverse risk factor in various studies. Recently, somatostatin receptor antagonists have been shown to be more sensitive and specific in detecting the disease. The aim of this review article is to summarize the clinical importance of SSTR-based imaging in the clinical management of neuroendocrine and related tumors.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
| |
Collapse
|
29
|
Grawe F, Rosenberger N, Ingenerf M, Beyer L, Eschbach R, Todica A, Seidensticker R, Schmid-Tannwald C, Cyran CC, Ricke J, Bartenstein P, Auernhammer CJ, Ruebenthaler J, Fabritius MP. Diagnostic performance of PET/CT in the detection of liver metastases in well-differentiated NETs. Cancer Imaging 2023; 23:41. [PMID: 37098632 PMCID: PMC10131442 DOI: 10.1186/s40644-023-00556-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/13/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND The aim of this retrospective study was to compare the diagnostic accuracy of somatostatin receptor (SSR)-PET/CT to liver MRI as reference standard in the evaluation of hepatic involvement in neuroendocrine tumors (NET). METHODS An institutional database was screened for "SSR" imaging studies between 2006 and 2021. 1000 NET Patients (grade 1/2) with 2383 SSR-PET/CT studies and matching liver MRI in an interval of +3 months were identified. Medical reports of SSR-PET/CT and MRI were retrospectively evaluated regarding hepatic involvement and either confirmed by both or observed in MRI but not in SSR-PET/CT (false-negative) or in SSR-PET but not in MRI (false-positive). RESULTS Metastatic hepatic involvement was reported in 1650 (69.2%) of the total 2383 SSR-PET/CT imaging studies, whereas MRI detected hepatic involvement in 1685 (70.7%) cases. There were 51 (2.1%) false-negative and 16 (0.7%) false-positive cases. In case of discrepant reports, MRI and PET/CT were reviewed side by side for consensus reading. SSR-PET/CT demonstrated a sensitivity of 97.0% (95%CI: 96.0%, 97.7%), a specificity of 97.7% (95%CI: 96.3%, 98.7%), a PPV of 99.0% (95%CI: 98.4%, 99.4%) and NPV of 93.0% (95%CI: 91.0, 94.8%) in identifying hepatic involvement. The most frequent reason for false-negative results was the small size of lesions with the majority < 0.6 cm. CONCLUSION This study confirms the high diagnostic accuracy of SSR-PET/CT in the detection of hepatic involvement in NET patients based on a patient-based analysis of metastatic hepatic involvement with a high sensitivity and specificity using liver MRI imaging as reference standard. However, one should be aware of possible pitfalls when a single imaging method is used in evaluating neuroendocrine liver metastases in patients.
Collapse
Affiliation(s)
- Freba Grawe
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
| | - Natalie Rosenberger
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Maria Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Ralf Eschbach
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
| | - Andrei Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Ricarda Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Christine Schmid-Tannwald
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Christoph J Auernhammer
- Department of Internal Medicine 4, University Hospital, LMU Munich, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Johannes Ruebenthaler
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM, ENETS certified Center of Excellence), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Matthias P Fabritius
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
30
|
[ 99mTc]Tc-HYNIC-RM2: A potential SPECT probe targeting GRPR expression in prostate cancers. Nucl Med Biol 2023; 118-119:108331. [PMID: 36933456 DOI: 10.1016/j.nucmedbio.2023.108331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023]
Abstract
INTRODUCTION Elevated density of gastrin releasing peptide receptors (GRPR) in prostate cancer has led to exploration of several radiolabeled peptides for imaging and staging of the disease. The GRPR antagonist peptide RM2 has been successfully conjugated with several chelators and radiolabeled with gallium-68. The goal of this study was to synthesize a 99mTc-labeled probe and investigate its potential for SPECT imaging of prostate cancer. Towards this HYNIC-RM2 peptide conjugate was synthesized, radiolabeled with 99mTc and evaluated in GRPR-positive PC3 tumor xenografts. METHODS HYNIC-RM2 was manually synthesized by standard Fmoc solid phase strategy and radiolabeled with 99mTc. In vitro cell studies were performed in GRPR-positive human prostate carcinoma (PC3) cells. Metabolic stability studies of [99mTc]Tc-HYNIC-RM2 were performed in normal mice in the presence as well as absence of neutral endopeptidase (NEP) inhibitor, phosphoramidon (PA). Biodistribution and imaging studies of [99mTc]Tc-HYNIC-RM2 were performed in SCID mice bearing PC3-xenograft. RESULTS [99mTc]Tc-HYNIC-RM2 exhibited high binding affinity in low nanomolar range (Kd = 1.83 ± 0.31 nM). Metabolic stability studies in mice indicated that in the absence of PA, radiolabeled peptide was about 65 % intact in the blood at 15 min p.i., whereas proportion of intact radiolabeled peptide was enhanced to 90 % on co-administration of PA. Biodistribution studies in PC3 tumor bearing mice demonstrated high tumor uptake (8.02 ± 0.9%ID/g and 6.13 ± 0.44%ID/g at 1 h and 3 h p.i.). Co-administration of PA with the radiolabeled peptide resulted in further enhancement of tumor uptake (14.24 ± 0.76 % ID/g and 11.71 ± 0.59%ID/g at 1 h and 3 h p.i.). SPECT/CT images of [99mTc]Tc-HYNIC-RM2 could clearly visualize the tumor. Significant (p < 0.001) reduction in the tumor uptake with a co-injected blocking dose of unlabeled peptide ascertained the GRPR specificity of [99mTc]Tc-HYNIC-RM2. CONCLUSION Encouraging results obtained in biodistribution and imaging studies indicate the potential of [99mTc]Tc-HYNIC-RM2 for further exploration as GRPR targeting agent.
Collapse
|
31
|
Correlations between [ 68Ga]Ga-DOTA-TOC Uptake and Absorbed Dose from [ 177Lu]Lu-DOTA-TATE. Cancers (Basel) 2023; 15:cancers15041134. [PMID: 36831477 PMCID: PMC9954147 DOI: 10.3390/cancers15041134] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE The aim of this paper was to investigate correlations between pre- therapeutic [68Ga]Ga-DOTA-TOC uptake and absorbed dose to tumours from therapy with [177Lu]Lu-DOTA-TATE. METHODS This retrospective study included 301 tumours from 54 GEP-NET patients. The tumours were segmented on pre-therapeutic [68Ga]Ga-DOTA-TOC PET/CT, and post-therapy [177Lu]Lu-DOTA-TATE SPECT/CT images, using a fixed 40% threshold. The SPECT/CT images were used for absorbed dose calculations by assuming a linear build-up from time zero to day one, and mono-exponential wash-out after that. Both SUVmean and SUVmax were measured from the PET images. A linear absorbed-dose prediction model was formed with SUVmean as the independent variable, and the accuracy was tested with a split 70-30 training-test set. RESULTS Mean SUVmean and SUVmax from [68Ga]Ga-DOTA-TOC PET was 24.0 (3.6-84.4) and 41.0 (6.7-146.5), and the mean absorbed dose from [177Lu]Lu-DOTA-TATE was 26.9 Gy (2.4-101.9). A linear relationship between SUVmean and [177Lu]Lu-DOTA-TATE activity concentration at 24 h post injection was found (R2 = 0.44, p < 0.05). In the prediction model, a root mean squared error and a mean absolute error of 1.77 and 1.33 Gy/GBq, respectively, were found for the test set. CONCLUSIONS There was a high inter- and intra-patient variability in tumour measurements, both for [68Ga]Ga-DOTA-TOC SUVs and absorbed doses from [177Lu]Lu-DOTA-TATE. Depending on the required accuracy, [68Ga]Ga-DOTA-TOC PET imaging may estimate the [177Lu]Lu-DOTA-TATE uptake. However, there could be a high variance between predicted and actual absorbed doses.
Collapse
|
32
|
Siebinga H, de Wit-van der Veen BJ, Beijnen JH, Dorlo TPC, Huitema ADR, Hendrikx JJMA. A physiologically based pharmacokinetic model for [ 68Ga]Ga-(HA-)DOTATATE to predict whole-body distribution and tumor sink effects in GEP-NET patients. EJNMMI Res 2023; 13:8. [PMID: 36735114 PMCID: PMC9898489 DOI: 10.1186/s13550-023-00958-7] [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: 03/25/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Little is known about parameters that have a relevant impact on (dis)similarities in biodistribution between various 68Ga-labeled somatostatin analogues. Additionally, the effect of tumor burden on organ uptake remains unclear. Therefore, the aim of this study was to describe and compare organ and tumor distribution of [68Ga]Ga-DOTATATE and [68Ga]Ga-HA-DOTATATE using a physiologically based pharmacokinetic (PBPK) model and to identify factors that might cause biodistribution and tumor uptake differences between both peptides. In addition, the effect of tumor burden on peptide biodistribution in gastroenteropancreatic (GEP) neuroendocrine tumor (NET) patients was assessed. METHODS A PBPK model was developed for [68Ga]Ga-(HA-)DOTATATE in GEP-NET patients. Three tumor compartments were added, representing primary tumor, liver metastases and other metastases. Furthermore, reactions describing receptor binding, internalization and recycling, renal clearance and intracellular degradation were added to the model. Scan data from GEP-NET patients were used for evaluation of model predictions. Simulations with increasing tumor volumes were performed to assess the tumor sink effect. RESULTS Data of 39 and 59 patients receiving [68Ga]Ga-DOTATATE and [68Ga]Ga-HA-DOTATATE, respectively, were included. Evaluations showed that the model adequately described image-based patient data and that different receptor affinities caused organ uptake dissimilarities between both peptides. Sensitivity analysis indicated that tumor blood flow and blood volume impacted tumor distribution most. Tumor sink predictions showed a decrease in spleen uptake with increasing tumor volume, which seemed clinically relevant for patients with total tumor volumes higher than ~ 550 mL. CONCLUSION The developed PBPK model adequately predicted tumor and organ uptake for this GEP-NET population. Relevant organ uptake differences between [68Ga]Ga-DOTATATE and [68Ga]Ga-HA-DOTATATE were caused by different affinity profiles, while tumor uptake was mainly affected by tumor blood flow and blood volume. Furthermore, tumor sink predictions showed that for the majority of patients a tumor sink effect is not expected to be clinically relevant.
Collapse
Affiliation(s)
- Hinke Siebinga
- grid.430814.a0000 0001 0674 1393Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.430814.a0000 0001 0674 1393Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Berlinda J. de Wit-van der Veen
- grid.430814.a0000 0001 0674 1393Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H. Beijnen
- grid.430814.a0000 0001 0674 1393Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thomas P. C. Dorlo
- grid.430814.a0000 0001 0674 1393Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.8993.b0000 0004 1936 9457Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Alwin D. R. Huitema
- grid.430814.a0000 0001 0674 1393Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.5477.10000000120346234Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands ,grid.487647.eDepartment of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jeroen J. M. A. Hendrikx
- grid.430814.a0000 0001 0674 1393Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands ,grid.430814.a0000 0001 0674 1393Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Patel R, Marano G, Joseph J, Chung J, Plata A, Vos JA. 18 F-fludeoxyglucose positron emission computed tomography ( 18 F-FDG-PET/CT) versus 68 Ga-DOTATATE-PET/CT in patients with head and neck cancer: Comparisons and implications for treatment. Head Neck 2023; 45:347-354. [PMID: 36591637 DOI: 10.1002/hed.27243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/22/2022] [Accepted: 10/28/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Tumor-specific molecular imaging in head and neck squamous cell carcinoma (HNSCC) is not well established. Somatostatin receptors (SSTRs) are found in solid tumors, including HNSCC. 68 Ga-DOTATATE, a commercially available radionuclide that binds SSTRs, may have utility in imaging HNSCC. METHODS Patients with HNSCC received pretreatment imaging with 18 F-FDG-PET/CT and 68 Ga-DOTATATE. Imaging was compared for concordance. When available, surgical resection specimens were compared to pretreatment imaging findings. Historic HNSCC tumor specimens were assessed for both SSTR and p16/human papilloma virus (HPV) expression. RESULTS Twenty patients were imaged. Fifteen had oropharyngeal cancer. Primary tumor site was concordant between imaging modalities for all patients. One of 45 lymph nodes was discordant. Retrospective specimen review showed a significant correlation with SSTR expression and HPV/p16 expression. No adverse events occurred. CONCLUSIONS 68 Ga-DOTATATE imaging is safe and effective in HNSCC. SSTR expression may be increased in HPV-mediated tumors. Targeted therapies to SSTR should be explored.
Collapse
Affiliation(s)
- Rusha Patel
- Department of Otolaryngology, Oklahoma University, Oklahoma City, Oklahoma, United States
| | - Gary Marano
- Department of Radiology, West Virginia University, Morgantown, West Virginia, USA
| | - Joe Joseph
- Department of Radiology, West Virginia University, Morgantown, West Virginia, USA
| | - Jeffson Chung
- Department of Otolaryngology, West Virginia University, Morgantown, West Virginia, USA
| | - Andrew Plata
- Department of Pathology, West Virginia University, Morgantown, West Virginia, USA
| | - Jeffrey A Vos
- Department of Pathology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
34
|
Pellegrino F, Granata V, Fusco R, Grassi F, Tafuto S, Perrucci L, Tralli G, Scaglione M. Diagnostic Management of Gastroenteropancreatic Neuroendocrine Neoplasms: Technique Optimization and Tips and Tricks for Radiologists. Tomography 2023; 9:217-246. [PMID: 36828370 PMCID: PMC9958666 DOI: 10.3390/tomography9010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) comprise a heterogeneous group of neoplasms, which derive from cells of the diffuse neuroendocrine system that specializes in producing hormones and neuropeptides and arise in most cases sporadically and, to a lesser extent, in the context of complex genetic syndromes. Furthermore, they are primarily nonfunctioning, while, in the case of insulinomas, gastrinomas, glucagonomas, vipomas, and somatostatinomas, they produce hormones responsible for clinical syndromes. The GEP-NEN tumor grade and cell differentiation may result in different clinical behaviors and prognoses, with grade one (G1) and grade two (G2) neuroendocrine tumors showing a more favorable outcome than grade three (G3) NET and neuroendocrine carcinoma. Two critical issues should be considered in the NEN diagnostic workup: first, the need to identify the presence of the tumor, and, second, to define the primary site and evaluate regional and distant metastases. Indeed, the primary site, stage, grade, and function are prognostic factors that the radiologist should evaluate to guide prognosis and management. The correct diagnostic management of the patient includes a combination of morphological and functional evaluations. Concerning morphological evaluations, according to the consensus guidelines of the European Neuroendocrine Tumor Society (ENETS), computed tomography (CT) with a contrast medium is recommended. Contrast-enhanced magnetic resonance imaging (MRI), including diffusion-weighted imaging (DWI), is usually indicated for use to evaluate the liver, pancreas, brain, and bones. Ultrasonography (US) is often helpful in the initial diagnosis of liver metastases, and contrast-enhanced ultrasound (CEUS) can solve problems in characterizing the liver, as this tool can guide the biopsy of liver lesions. In addition, intraoperative ultrasound is an effective tool during surgical procedures. Positron emission tomography (PET-CT) with FDG for nonfunctioning lesions and somatostatin analogs for functional lesions are very useful for identifying and evaluating metabolic receptors. The detection of heterogeneity in somatostatin receptor (SSTR) expression is also crucial for treatment decision making. In this narrative review, we have described the role of morphological and functional imaging tools in the assessment of GEP-NENs according to current major guidelines.
Collapse
Affiliation(s)
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Naples, Italy
| | - Francesca Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, 80127 Naples, Italy
| | - Salvatore Tafuto
- S.C. Sarcomi e Tumori Rari, Istituto Nazionale Tumori, IRCCS, Fondazione “G. Pascale”, 80131 Naples, Italy
| | - Luca Perrucci
- Ferrara Department of Interventional and Diagnostic Radiology, Ospedale di Lagosanto, Azienda AUSL, 44023 Ferrara, Italy
| | - Giulia Tralli
- Department of Radiology, Ospedale Santa Maria della Misericordia, 45100 Rovigo, Italy
| | - Mariano Scaglione
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
35
|
Shi L, Zhang J, Toyonaga T, Shao D, Onofrey JA, Lu Y. Deep learning-based attenuation map generation with simultaneously reconstructed PET activity and attenuation and low-dose application. Phys Med Biol 2023; 68. [PMID: 36584395 DOI: 10.1088/1361-6560/acaf49] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/30/2022] [Indexed: 12/31/2022]
Abstract
Objective. In PET/CT imaging, CT is used for positron emission tomography (PET) attenuation correction (AC). CT artifacts or misalignment between PET and CT can cause AC artifacts and quantification errors in PET. Simultaneous reconstruction (MLAA) of PET activity (λ-MLAA) and attenuation (μ-MLAA) maps was proposed to solve those issues using the time-of-flight PET raw data only. However,λ-MLAA still suffers from quantification error as compared to reconstruction using the gold-standard CT-based attenuation map (μ-CT). Recently, a deep learning (DL)-based framework was proposed to improve MLAA by predictingμ-DL fromλ-MLAA andμ-MLAA using an image domain loss function (IM-loss). However, IM-loss does not directly measure the AC errors according to the PET attenuation physics. Our preliminary studies showed that an additional physics-based loss function can lead to more accurate PET AC. The main objective of this study is to optimize the attenuation map generation framework for clinical full-dose18F-FDG studies. We also investigate the effectiveness of the optimized network on predicting attenuation maps for synthetic low-dose oncological PET studies.Approach. We optimized the proposed DL framework by applying different preprocessing steps and hyperparameter optimization, including patch size, weights of the loss terms and number of angles in the projection-domain loss term. The optimization was performed based on 100 skull-to-toe18F-FDG PET/CT scans with minimal misalignment. The optimized framework was further evaluated on 85 clinical full-dose neck-to-thigh18F-FDG cancer datasets as well as synthetic low-dose studies with only 10% of the full-dose raw data.Main results. Clinical evaluation of tumor quantification as well as physics-based figure-of-merit metric evaluation validated the promising performance of our proposed method. For both full-dose and low-dose studies, the proposed framework achieved <1% error in tumor standardized uptake value measures.Significance. It is of great clinical interest to achieve CT-less PET reconstruction, especially for low-dose PET studies.
Collapse
Affiliation(s)
- Luyao Shi
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
| | - Jiazhen Zhang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States of America
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States of America
| | - Dan Shao
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States of America.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China
| | - John A Onofrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America.,Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States of America.,Department of Urology, Yale University, New Haven, CT, United States of America
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States of America
| |
Collapse
|
36
|
Gherghe M, Lazar AM, Simion L, Irimescu IN, Sterea MC, Mutuleanu MD, Anghel RM. Standardized Uptake Values on SPECT/CT: A Promising Alternative Tool for Treatment Evaluation and Prognosis of Metastatic Neuroendocrine Tumours. Diagnostics (Basel) 2023; 13:318. [PMID: 36673128 PMCID: PMC9857822 DOI: 10.3390/diagnostics13020318] [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: 11/25/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
(1) Background: The aim of our study was to assess the feasibility of 99mTcEDDA/HYNIC-TOC SPECT/CT quantitative analysis in evaluating treatment response and disease progression in patients with NETs. (2) Methods: This prospective monocentric study evaluated 35 SPECT/CT examinations performed on 14 patients with neuroendocrine tumours who underwent a baseline and at least one follow-up 99mTcEDDA/HYNIC-TOC scan as part of their clinical management. The examination protocol included a whole-body scan acquired 2 h after the radiotracer’s administration, with the SPECT/CT performed 4 h post-injection. Images were analyzed by two experienced physicians and patients were classified into response categories based on their changes in SUV values. (3) Results: We evaluated 14 baseline studies and 21 follow-up scans, accounting for 123 lesions. A statistically positive correlation has been found between the SUVmax and SUVpeak values in tumoral lesions (p < 0.05). No correlation has been found between the SUV values and the ki67 proliferation index. Finally, 64.29% patients were classified as SD at the end of the study, with only 14.29% of patients exhibiting PD and 21.43% patients with PR. (4) Conclusions: The quantitative analysis of 99mTcEDDA/HYNIC-TOC SPECT/CT data in patients with neuroendocrine tumours could represent an alternative to 68Ga-DOTA-peptides PET/CT for the monitoring and prognosis of NETs.
Collapse
Affiliation(s)
- Mirela Gherghe
- Nuclear Medicine Department, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
- Nuclear Medicine Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Alexandra Maria Lazar
- Nuclear Medicine Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Laurentiu Simion
- Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- 1st Clinic of General Surgery and Surgical Oncology, Bucharest Oncology Institute, 022328 Bucharest, Romania
| | - Ionela-Nicoleta Irimescu
- Nuclear Medicine Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Maria-Carla Sterea
- Nuclear Medicine Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Mario-Demian Mutuleanu
- Nuclear Medicine Department, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
- Nuclear Medicine Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Oncology Department, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
- Radiotherapy Department, Institute of Oncology “Professor Doctor Alexandru Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
37
|
Al[ 18F]F-NOTA-Octreotide Is Comparable to [ 68Ga]Ga-DOTA-TATE for PET/CT Imaging of Neuroendocrine Tumours in the Latin-American Population. Cancers (Basel) 2023; 15:cancers15020439. [PMID: 36672388 PMCID: PMC9856643 DOI: 10.3390/cancers15020439] [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: 12/14/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
PET imaging of neuroendocrine tumours (NET) is well established for staging and therapy follow-up. The short half-life, increasing costs, and regulatory issues significantly limit the availability of approved imaging agents, such as [68Ga]Ga-DOTA-TATE. Al[18F]F-NOTA-Octreotide provides a similar biodistribution and tumour uptake, can be produced on a large scale and may improve access to precision imaging. Here we prospectively compared the clinical utility of [68Ga]Ga-DOTA-TATE and Al[18F]F-NOTA-Octreotide in the Latin-American population. Our results showed that in patients with stage IV NETs [68Ga]Ga-DOTA-TATE presents higher physiological uptake than Al[18F]F-NOTA-Octreotide in the liver, hypophysis, salivary glands, adrenal glands (all p < 0.001), pancreatic uncinated process, kidneys, and small intestine (all p < 0.05). Nevertheless, despite the lower background uptake of Al[18F]F-NOTA-Octreotide, comparative analysis of tumour-to-liver (TLR) and tumour-to-spleen (TSR) showed no statistically significant difference for lesions in the liver, bone, lymph nodes, and other tissues. Only three discordant lesions in highly-metastases livers were detected by [68Ga]Ga-DOTA-TATE but not by Al[18F]F-NOTA-Octreotide and only one discordant lesion was detected by Al[18F]F-NOTA-Octreotide but not by [68Ga]Ga-DOTA-TATE. Non-inferiority analysis showed that Al[18F]F-NOTA-Octreotide is comparable to [68Ga]Ga-DOTA-TATE. Hence, our results demonstrate that Al[18F]F-NOTA-Octreotide provided excellent image quality, visualized NET lesions with high sensitivity and represents a highly promising, clinical alternative to [68Ga]Ga-DOTA-TATE.
Collapse
|
38
|
Ouvrard E, Mestier LD, Boursier C, Lachachi B, Sahakian N, Chevalier E, Mikail N, Carullo J, Bando-Delaunay A, Walter T, Malouf GG, Addeo P, Poncet G, Sebag F, Lebtahi R, Goichot B, Taïeb D, Imperiale A. 18F-DOPA PET/CT at the Forefront of Initial or Presurgical Evaluation of Small-Intestine Neuroendocrine Tumors. J Nucl Med 2022; 63:1865-1870. [PMID: 35589408 DOI: 10.2967/jnumed.122.263984] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/27/2022] [Indexed: 01/11/2023] Open
Abstract
Our objective was to compare the respective value of 68Ga-DOTATOC and 18F-DOPA PET/CT for initial staging or presurgical work-up of patients with small-intestine neuroendocrine tumors (SiNETs). Methods: This was a retrospective, multicenter, noninterventional investigation involving 53 non-surgically treated SiNET patients who underwent both 68Ga-DOTATOC and 18F-DOPA PET/CT within a 6-mo interval without surgical intervention or therapeutic change between the 2 PET/CT studies. Percentage detection rate was calculated according to per-region and per-lesion analyses. Sensitivity for primary tumor detection was assessed in 37 surgically treated patients, taking surgical results (76 SiNETs) as the diagnostic gold standard. Results: 68Ga-DOTATOC PET/CT and 18F-DOPA PET/CT individually identified at least 1 primary SiNET in 92% (34/37) of the patients. Intestinal tumor multifocality was confirmed by histology in 8 patients. 68Ga-DOTATOC and 18F-DOPA PET/CT were concordantly positive for tumor multifocality in 5 patients, discordantly positive in 2 patients, and concordantly negative in 1 patient. The detection rate for subdiaphragmatic nodal metastases on per-region-based analysis was 91% and 98% for 68Ga-DOTATOC and 18F-DOPA PET/CT, respectively (P = 0.18). 18F-DOPA PET/CT detected a higher number of abnormal subdiaphragmatic nodes (P = 0.009). Regarding mesenteric nodes only, 18F-DOPA PET/CT detected more positive regions (P = 0.005) and nodal lesions (P = 0.003) than 68Ga-DOTATOC PET/CT, including nodes at the origin of mesenteric vessels. For detection of distant metastases, 68Ga-DOTATOC and 18F-DOPA PET/CT performed equally well on a per-region-based analysis. As compared with 68Ga-DOTATOC, 18F-DOPA PET/CT detected more hepatic (P < 0.001), peritoneal (P < 0.001), and lung metastases (P < 0.001). Conclusion: 18F-DOPA PET/CT detected more lesions than 68Ga-DOTATOC PET/CT in the studied patients. The respective roles of the two should be discussed in terms of disease staging and treatment selection.
Collapse
Affiliation(s)
- Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe, University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Louis De Mestier
- Department of Pancreatology and Digestive Oncology, ENETS Centre of Excellence, Beaujon Hospital, Université de Paris, and INSERM U1149, Centre of Research in Inflammation, Paris, France
| | | | | | - Nicolas Sahakian
- Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France
| | | | - Nidaa Mikail
- Nuclear Medicine, ENETS Centre of Excellence, Beaujon Hospital, Université de Paris, Paris, France
| | - Josefina Carullo
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe, University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France.,Nuclear Medicine, Sanatorio Allende S.A., Cordoba, Argentina
| | - Aurélie Bando-Delaunay
- Nuclear Medicine, ENETS Centre of Excellence, Beaujon Hospital, Université de Paris, Paris, France
| | - Thomas Walter
- Medical Oncology, Edouard Herriot Hospital, Hospices Civils de Lyon, Université Lyon 1, Lyon, France
| | - Gabriel G Malouf
- Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Pietro Addeo
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Hospitals of Strasbourg, Strasbourg, France
| | - Gilles Poncet
- Digestive and Oncologic Surgery, Edouard-Herriot University Hospital, Claude-Bernard Lyon 1 University, Lyon, France
| | - Frederic Sebag
- Endocrine Surgery, Conception University Hospital, Aix-Marseille University, Marseille, France
| | - Rachida Lebtahi
- Nuclear Medicine, ENETS Centre of Excellence, Beaujon Hospital, Université de Paris, Paris, France
| | - Bernard Goichot
- Diabetes and Metabolic Disorders, Internal Medicine, University Hospitals of Strasbourg, Strasbourg University, Strasbourg, France
| | - David Taïeb
- Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France.,European Center for Research in Medical Imaging, Aix-Marseille University, Marseille, France; and
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe, University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France; .,Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| |
Collapse
|
39
|
Prosperi D, Gentiloni Silveri G, Panzuto F, Faggiano A, Russo VM, Caruso D, Polici M, Lauri C, Filice A, Laghi A, Signore A. Nuclear Medicine and Radiological Imaging of Pancreatic Neuroendocrine Neoplasms: A Multidisciplinary Update. J Clin Med 2022; 11:jcm11226836. [PMID: 36431313 PMCID: PMC9694730 DOI: 10.3390/jcm11226836] [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: 10/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are part of a large family of tumors arising from the neuroendocrine system. PanNENs show low-intermediate tumor grade and generally high somatostatin receptor (SSTR) expression. Therefore, panNENs benefit from functional imaging with 68Ga-somatostatin analogues (SSA) for diagnosis, staging, and treatment choice in parallel with morphological imaging. This narrative review aims to present conventional imaging techniques and new perspectives in the management of panNENs, providing the clinicians with useful insight for clinical practice. The 68Ga-SSA PET/CT is the most widely used in panNENs, not only fr diagnosis and staging purpose but also to characterize the biology of the tumor and its responsiveness to SSAs. On the contrary, the 18F-Fluordeoxiglucose (FDG) PET/CT is not employed systematically in all panNEN patients, being generally preferred in G2-G3, to predict aggressiveness and progression rate. The combination of 68Ga-SSA PET/CT and 18F-FDG PET/CT can finally suggest the best therapeutic strategy. Other radiopharmaceuticals are 68Ga-exendin-4 in case of insulinomas and 18F-dopamine (DOPA), which can be helpful in SSTR-negative tumors. New promising but still-under-investigation radiopharmaceuticals include radiolabeled SSTR antagonists and 18F-SSAs. Conventional imaging includes contrast enhanced CT and multiparametric MRI. There are now enriched by radiomics, a new non-invasive imaging approach, very promising to early predict tumor response or progression.
Collapse
Affiliation(s)
- Daniela Prosperi
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Guido Gentiloni Silveri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Department of Medical-Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Vincenzo Marcello Russo
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Michela Polici
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Chiara Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
- Correspondence:
| | - Angelina Filice
- Nucler Medicine Unit, AUSL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Laghi
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| |
Collapse
|
40
|
Evaluation of 68Ga-Radiolabeled Peptides for HER2 PET Imaging. Diagnostics (Basel) 2022; 12:diagnostics12112710. [PMID: 36359554 PMCID: PMC9689602 DOI: 10.3390/diagnostics12112710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
One in eight women will be diagnosed with breast cancer in their lifetime and approximately 25% of those cases will be HER2-positive. Current methods for diagnosing HER2-positive breast cancer involve using IHC and FISH from suspected cancer biopsies to quantify HER2 expression. HER2 PET imaging could potentially increase accuracy and improve the diagnosis of lesions that are not available for biopsies. Using two previously discovered HER2-targeting peptides, we modified each peptide with the chelator DOTA and a PEG2 linker resulting in DOTA-PEG2-GSGKCCYSL (P5) and DOTA-PEG2-DTFPYLGWWNPNEYRY (P6). Each peptide was labeled with 68Ga and was evaluated for HER2 binding using in vitro cell studies and in vivo tumor xenograft models. Both [68Ga]P5 and [68Ga]P6 showed significant binding to HER2-positive BT474 cells versus HER2-negative MDA-MB-231 cells ([68Ga]P5; 0.68 ± 0.20 versus 0.47 ± 0.05 p < 0.05 and [68Ga]P6; 0.55 ± 0.21 versus 0.34 ± 0.12 p < 0.01). [68Ga]P5 showed a higher percent injected dose per gram (%ID/g) binding to HER2-positive tumors two hours post-injection compared to HER2-negative tumors (0.24 ± 0.04 versus 0.12 ± 0.06; p < 0.05), while the [68Ga]P6 peptide showed significant binding (0.98 ± 0.22 versus 0.51 ± 0.08; p < 0.05) one hour post-injection. These results lay the groundwork for the use of peptides to image HER2-positive breast cancer.
Collapse
|
41
|
Lin M, Coll RP, Cohen AS, Georgiou DK, Manning HC. PET Oncological Radiopharmaceuticals: Current Status and Perspectives. Molecules 2022; 27:6790. [PMID: 36296381 PMCID: PMC9609795 DOI: 10.3390/molecules27206790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 02/01/2024] Open
Abstract
Molecular imaging is the visual representation of biological processes that take place at the cellular or molecular level in living organisms. To date, molecular imaging plays an important role in the transition from conventional medical practice to precision medicine. Among all imaging modalities, positron emission tomography (PET) has great advantages in sensitivity and the ability to obtain absolute imaging quantification after corrections for photon attenuation and scattering. Due to the ability to label a host of unique molecules of biological interest, including endogenous, naturally occurring substrates and drug-like compounds, the role of PET has been well established in the field of molecular imaging. In this article, we provide an overview of the recent advances in the development of PET radiopharmaceuticals and their clinical applications in oncology.
Collapse
Affiliation(s)
- Mai Lin
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ryan P. Coll
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Allison S. Cohen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dimitra K. Georgiou
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Henry Charles Manning
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
42
|
Park J, Young BD, Miller EJ. Potential novel imaging targets of inflammation in cardiac sarcoidosis. J Nucl Cardiol 2022; 29:2171-2187. [PMID: 34734365 DOI: 10.1007/s12350-021-02838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/26/2021] [Indexed: 10/19/2022]
Abstract
Cardiac sarcoidosis (CS) is an inflammatory disease with high morbidity and mortality, with a pathognomonic feature of non-caseating granulomatous inflammation. While 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) is a well-established modality to image inflammation and diagnose CS, there are limitations to its specificity and reproducibility. Imaging focused on the molecular processes of inflammation including the receptors and cellular microenvironments present in sarcoid granulomas provides opportunities to improve upon FDG-PET imaging for CS. This review will highlight the current limitations of FDG-PET imaging for CS while discussing emerging new nuclear imaging molecular targets for the imaging of cardiac sarcoidosis.
Collapse
Affiliation(s)
- Jakob Park
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Bryan D Young
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
43
|
McClellan K, Chen EY, Kardosh A, Lopez CD, Del Rivero J, Mallak N, Rocha FG, Koethe Y, Pommier R, Mittra E, Pegna GJ. Therapy Resistant Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2022; 14:4769. [PMID: 36230691 PMCID: PMC9563314 DOI: 10.3390/cancers14194769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are a heterogenous group of malignancies originating from neuroendocrine cells of the gastrointestinal tract, the incidence of which has been increasing for several decades. While there has been significant progress in the development of therapeutic options for patients with advanced or metastatic disease, these remain limited both in quantity and durability of benefit. This review examines the latest research elucidating the mechanisms of both up-front resistance and the eventual development of resistance to the primary systemic therapeutic options including somatostatin analogues, peptide receptor radionuclide therapy with lutetium Lu 177 dotatate, everolimus, sunitinib, and temozolomide-based chemotherapy. Further, potential strategies for overcoming these mechanisms of resistance are reviewed in addition to a comprehensive review of ongoing and planned clinical trials addressing this important challenge.
Collapse
Affiliation(s)
- Kristen McClellan
- School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Emerson Y. Chen
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Adel Kardosh
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Charles D. Lopez
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nadine Mallak
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR 97239, USA
| | - Flavio G. Rocha
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yilun Koethe
- Dotter Department of Interventional Radiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rodney Pommier
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Erik Mittra
- Division of Molecular Imaging and Therapy, Oregon Health & Science University, Portland, OR 97239, USA
| | - Guillaume J. Pegna
- Division of Hematology Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
44
|
Lee ONY, Tan KV, Tripathi V, Yuan H, Chan WWL, Chiu KWH. The Role of 68 Ga-DOTA-SSA PET/CT in the Management and Prediction of Peptide Receptor Radionuclide Therapy Response for Patients With Neuroendocrine Tumors : A Systematic Review and Meta-analysis. Clin Nucl Med 2022; 47:781-793. [PMID: 35485851 DOI: 10.1097/rlu.0000000000004235] [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: 11/25/2022]
Abstract
PURPOSE The aim of this study was to identify and evaluate the role of 68 Ga-DOTA-somatostatin analog (SSA) PET/CT in guiding treatment for patients with neuroendocrine tumors (NETs) based on published literature, with specific focus on the ability of PET/CT to impact clinical management and predict peptide receptor radionuclide therapy (PRRT) response. PATIENTS AND METHODS A systematic literature search of articles up to December 2021 was performed using PubMed and Scopus. Eligible studies included ≥10 patients with confirmed or suspected NETs who had undergone pretreatment staging 68 Ga-DOTA-SSA PET/CT. A meta-analysis using the random-effects model was conducted to determine the overall change in management after PET/CT, whereas PET/CT-derived parameters that correlated with PRRT outcome were summarized from studies that assessed its predictive capabilities. RESULTS A total of 39 studies were included in this systemic review, of which 2266 patients from 24 studies were included for meta-analysis. We showed that PET/CT resulted in a change in clinical management in 36% (95% confidence interval, 31%-41%; range, 3%-66%) of patients. Fifteen studies consisting of 618 patients examined the prognostic ability of 68 Ga-DOTA-SSA PET/CT for PRRT. Of those, 8 studies identified a higher pretreatment SUV to favor PRRT, and 4 identified PET-based radiomic features for somatostatin receptor heterogeneity to be predictive of PRRT response. CONCLUSIONS Along with its diagnostic abilities, 68 Ga-DOTA-SSA PET/CT can impact treatment decision-making and may predict PRRT response in patients with NETs. More robust studies should be conducted to better elucidate the prognostic role of somatostatin receptor PET/CT in optimizing treatment for clinical outcome.
Collapse
Affiliation(s)
- Osher Ngo Yung Lee
- From the Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kel Vin Tan
- Department of Oncology, The University of Oxford, Oxford, United Kingdom
| | - Vrijesh Tripathi
- Department of Mathematics and Statistics, The University of the West Indies, St. Augustine Campus, Trinidad and Tobago
| | - Hui Yuan
- Department of Nuclear Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Keith Wan Hang Chiu
- Department of Diagnostic and Interventional Radiology, Kwong Wah Hospital, Hong Kong
| |
Collapse
|
45
|
Kolade OU, Ayeni AO, Brink A, Steyn R, More S, Prasad V. SARS-CoV-2 vaccination site as possible pitfall on somatostatin receptor imaging. Clin Transl Imaging 2022; 10:579-585. [PMID: 35968530 PMCID: PMC9362710 DOI: 10.1007/s40336-022-00519-3] [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: 06/04/2022] [Accepted: 07/25/2022] [Indexed: 11/27/2022]
Abstract
SARS-CoV-2 (COVID-19) vaccination numbers are globally increasing. Therefore, an increased chance exists that patients undergoing Peptide Receptor Radionuclide Therapy (PRRT) or diagnostic radionuclide imaging for Neuroendocrine Tumours (NETs) may have recently received vaccination. We report the imaging findings of two NETs patients, A—following [177Lu] Lu-DOTATATE PRRT post therapy planar scintigraphy and single photon emission computed tomography with computed tomography (SPECT/CT), and B—following [68 Ga]Ga-DOTA-NOC positron emission tomography with computed tomography (PET/CT) respectively. Both studies were done few days after COVID-19 vaccination. Patient A showed a new focus of uptake in the left deltoid muscle; and Patient B showed uptake in the left deltoid and a left axillary lymph node. Nuclear Physicians need to be aware of pitfalls with somatostatin receptor radionuclide imaging post-vaccination to ensure accurate interpretation, as well as dosimetric considerations with vaccine-related post-therapy uptake.
Collapse
Affiliation(s)
- Olumayowa U. Kolade
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
- Division of Nuclear Medicine, Department of Radiation Medicine, Groote Schuur Hospital, University of Cape Town, C3/4 New Main Building, Cape Town, South Africa
| | - Akinwale O. Ayeni
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
| | - Anita Brink
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
| | - Rachelle Steyn
- Department of Nuclear Medicine, Hawke’s Bay District Health Board, Hastings, New Zealand
| | - Stuart More
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
| | - Vikas Prasad
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
46
|
Somatostatin Receptor Targeted PET-Imaging for Diagnosis, Radiotherapy Planning and Theranostics of Meningiomas: A Systematic Review of the Literature. Diagnostics (Basel) 2022; 12:diagnostics12071666. [PMID: 35885570 PMCID: PMC9321668 DOI: 10.3390/diagnostics12071666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
The aims of the present systematic review are to: (1) assess the diagnostic performance of somatostatin receptor (SSR)targeted positron emission tomography (PET) with different tracers and devices in patients affected by meningiomas; and (2) to evaluate the theranostic applications of peptide receptor radionuclide therapy (PRRT) in meningiomas. A systematic literature search according to PRISMA criteria was made by using two main databases. Only studies published from 2011 up to March 2022 in the English language with ≥10 enrolled patients were selected. Following our research strategy, 17 studies were included for the assessment. Fourteen studies encompassed 534 patients, harboring 733 meningiomas, submitted to SSR-targeted PET/CT (n = 10) or PET/MRI (n = 4) for de novo diagnosis, recurrence detection, or radiation therapy (RT) planning (endpoint 1), while 3 studies included 69 patients with therapy-refractory meningiomas submitted to PRRT (endpoint 2). A relevant variation in methodology was registered among diagnostic studies, since only a minority of them reported histopathology as a reference standard. PET, especially when performed through PET/MRI, resulted particularly useful for the detection of meningiomas located in the skull base (SB) or next to the falx cerebri, significantly influencing RT planning. As far as it concerns PRRT studies, stable disease was obtained in the 66.6% of the treated patients, being grade 1–2 hematological toxicity the most common side effect. Of note, the wide range of the administered activities, the various utilized radiopharmaceuticals (90Y-DOTATOC and/or 177Lu-DOTATATE), the lack of dosimetric studies hamper a clear definition of PRRT potential on meningiomas’ management.
Collapse
|
47
|
Toyonaga T, Shao D, Shi L, Zhang J, Revilla EM, Menard D, Ankrah J, Hirata K, Chen MK, Onofrey JA, Lu Y. Deep learning-based attenuation correction for whole-body PET - a multi-tracer study with 18F-FDG, 68 Ga-DOTATATE, and 18F-Fluciclovine. Eur J Nucl Med Mol Imaging 2022; 49:3086-3097. [PMID: 35277742 PMCID: PMC10725742 DOI: 10.1007/s00259-022-05748-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/25/2022] [Indexed: 11/04/2022]
Abstract
A novel deep learning (DL)-based attenuation correction (AC) framework was applied to clinical whole-body oncology studies using 18F-FDG, 68 Ga-DOTATATE, and 18F-Fluciclovine. The framework used activity (λ-MLAA) and attenuation (µ-MLAA) maps estimated by the maximum likelihood reconstruction of activity and attenuation (MLAA) algorithm as inputs to a modified U-net neural network with a novel imaging physics-based loss function to learn a CT-derived attenuation map (µ-CT). METHODS Clinical whole-body PET/CT datasets of 18F-FDG (N = 113), 68 Ga-DOTATATE (N = 76), and 18F-Fluciclovine (N = 90) were used to train and test tracer-specific neural networks. For each tracer, forty subjects were used to train the neural network to predict attenuation maps (µ-DL). µ-DL and µ-MLAA were compared to the gold-standard µ-CT. PET images reconstructed using the OSEM algorithm with µ-DL (OSEMDL) and µ-MLAA (OSEMMLAA) were compared to the CT-based reconstruction (OSEMCT). Tumor regions of interest were segmented by two radiologists and tumor SUV and volume measures were reported, as well as evaluation using conventional image analysis metrics. RESULTS µ-DL yielded high resolution and fine detail recovery of the attenuation map, which was superior in quality as compared to µ-MLAA in all metrics for all tracers. Using OSEMCT as the gold-standard, OSEMDL provided more accurate tumor quantification than OSEMMLAA for all three tracers, e.g., error in SUVmax for OSEMMLAA vs. OSEMDL: - 3.6 ± 4.4% vs. - 1.7 ± 4.5% for 18F-FDG (N = 152), - 4.3 ± 5.1% vs. 0.4 ± 2.8% for 68 Ga-DOTATATE (N = 70), and - 7.3 ± 2.9% vs. - 2.8 ± 2.3% for 18F-Fluciclovine (N = 44). OSEMDL also yielded more accurate tumor volume measures than OSEMMLAA, i.e., - 8.4 ± 14.5% (OSEMMLAA) vs. - 3.0 ± 15.0% for 18F-FDG, - 14.1 ± 19.7% vs. 1.8 ± 11.6% for 68 Ga-DOTATATE, and - 15.9 ± 9.1% vs. - 6.4 ± 6.4% for 18F-Fluciclovine. CONCLUSIONS The proposed framework provides accurate and robust attenuation correction for whole-body 18F-FDG, 68 Ga-DOTATATE and 18F-Fluciclovine in tumor SUV measures as well as tumor volume estimation. The proposed method provides clinically equivalent quality as compared to CT in attenuation correction for the three tracers.
Collapse
Affiliation(s)
- Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Dan Shao
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Guangdong Provincial People's Hospital, Guangzhou, Guangdong, China
| | - Luyao Shi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Jiazhen Zhang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Enette Mae Revilla
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | | | | | - Kenji Hirata
- Department of Diagnostic Imaging, School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Yale New Haven Hospital, New Haven, CT, USA
| | - John A Onofrey
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Urology, Yale University, New Haven, CT, USA
| | - Yihuan Lu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
| |
Collapse
|
48
|
Zandee WT, Merola E, Poczkaj K, de Mestier L, Klümpen HJ, Geboes K, de Herder WW, Munir A. Evaluation of multidisciplinary team decisions in neuroendocrine neoplasms: Impact of expert centres. Eur J Cancer Care (Engl) 2022; 31:e13639. [PMID: 35735226 DOI: 10.1111/ecc.13639] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/10/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate the impact of multidisciplinary team (MDT) meetings on the management of patients with neuroendocrine neoplasms (NENs). METHODS All newly referred gastro-entero-pancreatic (GEP)-NEN patients discussed from 1 April to 1 October 2017 in the MDT of seven European expert centres were prospectively included. The impact on patients' management was defined as a change in diagnosis, grade, stage or treatment. RESULTS A total of 292 patients were included, mainly small intestinal (siNENs) (32%) and pancreatic NENs (28%), with distant metastases in 51%. Patients had received prior surgery in 43% of cases and prior medical treatment in 32%. A significant change occurred in 61% of NENs: 7% changes in diagnosis, 8% in grade and 16% in stage. The MDT recommended a new treatment for 51% of patients, mainly surgery (9%) or somatostatin analogues (20%). A significant change was most frequently observed in patients with Stage IV disease (hazard ratio [HR] 3.6, 95% confidence interval [CI]: 1.9-6.9 vs. Stage I) and G2 NENs (vs. G1, HR 2.1 95% CI: 1.2-3.8). CONCLUSION NEN-dedicated MDT discussion in expert centres yields significant management changes in over 60% of patients and thus represents the gold standard for the management of these patients.
Collapse
Affiliation(s)
- Wouter T Zandee
- Department of Internal Medicine, Sector of Endocrinology Erasmus MC, Rotterdam, The Netherlands.,Department of Internal Medicine, Division of Endocrinology, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - Elettra Merola
- Department of Medicine 1, Division of Endocrinology, Friedrich-Alexander University, Erlangen-Nuremberg, Erlangen, Germany.,Department of Gastroenterology, Azienda Provinciale Servizi Sanitari (APSS), Trento, Italy
| | - Karolina Poczkaj
- Department of Endocrinology and Neuroendocrine Tumours, Medical University of Silesia, Katowice, Poland
| | - Louis de Mestier
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (APHP) and Paris 7 University, Clichy, France
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Karen Geboes
- Department of Gastroenterology, University Hospital Ghent, Ghent, Belgium
| | - Wouter W de Herder
- Department of Internal Medicine, Sector of Endocrinology Erasmus MC, Rotterdam, The Netherlands
| | - Alia Munir
- Department of Endocrinology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
49
|
Jiang Y, Liu Q, Wang G, Sui H, Wang R, Wang J, Zhu Z. A prospective head-to-head comparison of 68 Ga-NOTA-3P-TATE-RGD and 68 Ga-DOTATATE in patients with gastroenteropancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2022; 49:4218-4227. [PMID: 35657429 DOI: 10.1007/s00259-022-05852-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of this study was to compare 68 Ga-NOTA-3P-TATE-RGD, a dual somatostatin receptor 2- and integrin αVβ3-targeting tracer, to 68 Ga-DOTATATE in a single group of patients with gastroenteropancreatic (GEP)-neuroendocrine tumours (NETs). METHODS Thirty-five patients with histologically confirmed GEP-NETs (5 grade 1, 28 grade 2, and 2 grade 3 tumours) were prospectively enrolled with informed consent. The primary tumour mainly originated from the pancreas and rectum. All patients were scanned with both 68 Ga-NOTA-3P-TATE-RGD PET/CT and 68 Ga-DOTATATE PET/CT within a week and compared on a head-to-head basis. Sixteen patients also had conventional 18F-FDG PET/CT. Images were evaluated semi-quantitatively using maximum standardized uptake values (SUVmax) of tumour and tumour-to-background ratio. RESULTS All patients had at least one positive lesion on each of the two scans. A total of 1190 and 1106 lesions were detected on 68 Ga-NOTA-3P-TATE-RGD images and 68 Ga-DOTATATE images, respectively (P = 0.152). 68 Ga-NOTA-3P-TATE-RGD PET/CT revealed significantly more lesions in the liver than 68 Ga-DOTATATE PET/CT (634 vs. 532, P = 0.021). Both tracers produced comparable results for detecting primary tumours (20 vs. 20, P = 1.000), lymph node metastases (101 vs. 102, P = 0.655), and bone metastases (381 vs. 398, P = 0.244). The tumour SUVmax in 12 patients was significantly higher for 68 Ga-NOTA-3P-TATE-RGD than for 68 Ga-DOTATATE (27.2 ± 13.6 vs. 19.5 ± 10.0, P < 0.001); among them, 9 had 18F-FDG PET/CT and all were found to be FDG-positive. The remaining 23 patients had significantly higher 68 Ga-DOTATATE uptake than 68 Ga-NOTA-3P-TATE-RGD uptake (22.3 ± 16.4 vs. 11.9 ± 7.5, P < 0.001); among them, 7 had 18F-FDG PET/CT and 6 were FDG-negative. Generally, 68 Ga-DOTATATE demonstrated higher tumour SUVmax than 68 Ga-NOTA-3P-TATE-RGD (20.8 ± 16.0 vs. 14.2 ± 8.9, P < 0.001), including primary tumours, liver lesions, lymph node lesions, and bone lesions. However, the tumour-to-background ratio of liver lesions was significantly higher when using 68 Ga-NOTA-3P-TATE-RGD compared with that when using 68 Ga-DOTATATE (8.4 ± 5.5 vs. 4.7 ± 3.7, P < 0.001). CONCLUSION 68 Ga-NOTA-3P-TATE-RGD performed better than 68 Ga-DOTATATE in detection of liver metastases with a higher tumour-to-background ratio. Moreover, 68 Ga-NOTA-3P-TATE-RGD tended to demonstrate higher uptake over 68 Ga-DOTATATE in FDG-avid NETs. TRIAL REGISTRATION Dual SSTR2 and Integrin αvβ3 Targeting PET/CT Imaging (NCT02817945, registered 5 November 2018). URL OF REGISTRY: https://clinicaltrials.gov/ct2/show/NCT02817945.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Qingxing Liu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Guochang Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Huimin Sui
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Rongxi Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Jiarou Wang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China. .,State Key Laboratory of Complex Severe and Rare Diseases, Beijing, 100730, China.
| |
Collapse
|
50
|
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
|