1
|
Jülke EM, Beck-Sickinger AG. Peptide therapeutics: current status and future opportunity with focus on nose-to-brain delivery☆. Peptides 2025; 188:171404. [PMID: 40222598 DOI: 10.1016/j.peptides.2025.171404] [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: 11/26/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Peptide drugs are a highly diverse group of therapeutic agents. Over the last decade, more than 40 peptides have been approved for clinical use. They target different structures, ranging from G protein-coupled receptors (GPCRs) to pathogens and are used to treat a variety of indications, including metabolic disorders, genetic diseases, acute illnesses and more. Structurally, peptide therapeutics are a heterogeneous class. This diversity allows them to bridge the gap between small molecules and biologics. However, limited metabolic stability and bioavailability must be addressed. Strategies to improve the half-life include backbone and sequence modification, cyclization and the addition of stabilizing moieties. Great strides have been made in recent years towards achieving sufficient drug uptake for oral application have been achieved within recent years. However, these methods require specialized peptide design or involve permeabilization of the gastrointestinal tract. Consequently, other routes of administration are being explored. One promising approach is the nasal application of peptides. This method can be used for systemic uptake, but also allows for direct nose-to-brain delivery of compounds. While successful nose-to-brain delivery is already used in the clinic, the underlining mechanisms are poorly understood. Strategies for rational optimization are needed to make this method more applicable to a wider range of compounds. Overall, approved peptide therapeutics cover a wide range of applications and have demonstrated a growing and novel potential in recent drug discovery.
Collapse
Affiliation(s)
- Eva-Maria Jülke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany.
| |
Collapse
|
2
|
Abdlkadir AS, Al-Adhami D, Al Rammahi M, Badarneh M, Al Yasjeen S, Al Busaidi K, Khalaf A, Al-Alawi H, Al-Alawi H, Al-Ibraheem A. Diagnostic pitfalls in [68Ga]Ga-DOTATATE PET/CT imaging: a systematic review. Nucl Med Commun 2025:00006231-990000000-00423. [PMID: 40325977 DOI: 10.1097/mnm.0000000000001987] [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: 05/07/2025]
Abstract
[68Ga]Ga-DOTA-Tyr3-octreotate ([68Ga]Ga-DOTATATE) is an established somatostatin receptor imaging agent that has demonstrated superior efficacy in visualizing neuroendocrine tumors (NETs) and meningiomas compared with traditional [111In]In-octreotide imaging. Despite its enhanced affinity and sensitivity, [68Ga]Ga-DOTATATE imaging is not without challenges. To date, numerous diagnostic pitfalls and false-positive findings have been reported. This systematic review investigates the currently recognized diagnostic pitfalls in [68Ga]Ga-DOTATATE positron imaging. A systematic literature search was conducted using PubMed, Scopus, and Web of Science databases, with the most recent update on 8 March 2024. Two authors screened the titles and abstracts of retrieved articles and selected studies based on predefined inclusion and exclusion criteria. Qualitative analysis of 70 included research articles, encompassing 199 patients, identified 234 diagnostic pitfalls. Malignant neoplastic etiologies predominated, constituting 56% of pitfalls, followed by nononcologic pitfalls (32.1%), and benign oncologic tumors (11.9%). Anatomically, the head and neck region was the most frequent site for pitfalls (35.5%), followed by the musculoskeletal system (27.4%), abdomen (17.5%), and chest (16.6%). Pelvic-related pitfalls were least common, accounting for only 3% of cases. This study details potential diagnostic pitfalls, predominantly occurring in the head-neck regions - primary sites for meningiomas and paragangliomas. Understanding these diagnostic pitfalls is crucial for accurate diagnosis. Moreover, recognizing these diagnostic pitfalls may lead to novel applications of [68Ga]Ga-DOTATATE beyond its conventional use in NETs and meningiomas, potentially expanding its diagnostic utility.
Collapse
Affiliation(s)
- Ahmed Saad Abdlkadir
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Dhuha Al-Adhami
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Mohammed Al Rammahi
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Mohannad Badarneh
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Salem Al Yasjeen
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Khalid Al Busaidi
- Department of Nuclear Medicine and Molecular Imaging, Royal Hospital, Muscat, Oman,
| | - Aysar Khalaf
- Department of Nuclear Medicine, Warith International Cancer Institute, Karbala,
| | - Haider Al-Alawi
- Department of Nuclear Medicine, Amir Al-Momineen Specialty Hospital, Najaf, Iraq and
| | - Hasan Al-Alawi
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
| | - Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Amman, Jordan,
- Division of Nuclear Medicine, Department of Radiology and Nuclear Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
3
|
Hou G, Cheng X, Jiang Y, Zheng R, Wang X. A Rare Case of Peritoneal Mesothelioma Detected by 18 F-AlF-NOTA-Octreotide PET/CT. Clin Nucl Med 2025; 50:446-447. [PMID: 39853210 DOI: 10.1097/rlu.0000000000005681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 01/26/2025]
Abstract
ABSTRACT We present a case of 37-year-old man with multiple masses in the abdominal and pelvic cavity who underwent 18 F-AlF-NOTA-octreotide PET/CT. The masses demonstrated heterogeneously increased uptake on 18 F-AlF-NOTA-octreotide PET/CT and were suggestive of neuroendocrine tumor. However, the histopathological examinations confirmed the masses to be peritoneal mesothelioma. This case indicated that peritoneal mesothelioma should be kept in mind as one of the differential diagnoses for 18 F-AlF-NOTA-octreotide-avid tumors.
Collapse
Affiliation(s)
- Guozhu Hou
- Department of Nuclear Medicine (PET-CT Center), National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | |
Collapse
|
4
|
Filizoglu N, Ozguven S, Kesim S, Oksuzoglu K, Caglıyan F, Ones T, Dede F, Turoglu HT, Erdil TY. Physiological bio-distribution of 68Ga-DOTA-TATE in pediatric patients. Ann Nucl Med 2025:10.1007/s12149-025-02040-9. [PMID: 40106206 DOI: 10.1007/s12149-025-02040-9] [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: 11/15/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Somatostatin receptors (SSTRs) are G protein-coupled transmembrane receptors that serve as a specific molecular target for a number of radiopharmaceuticals utilized for the imaging of neuroendocrine tumors (NETs). 68Ga-DOTA-TATE is a somatostatin analog that demonstrates a high affinity for SSTR2. Pediatric malignancies, such as neuroblastoma, pheochromocytoma, and paraganglioma, have been shown to express SSTR2, and 68Ga-DOTA-TATE is currently being used to evaluate these pediatric neoplasms. We aimed to analyze the distribution pattern of 68Ga-DOTA-TATE based on age and location in pediatric patients. METHODS We retrospectively analyzed 247 consecutive 68Ga-DOTA-TATE whole-body PET/CT scans performed in our department from May 2015 to April 2024 in pediatric patients with known or suspected neuroblastoma, neuroendocrine malignancy, pheochromocytoma, and paraganglioma. 93 subjects were included in this study who were disease-free at the time of imaging and had no tracer-avid lesion on 68Ga-DOTA-TATE PET/CT. The patients were divided into four groups according to age: infant (0-2 years), pre-school (3-6 years), school (7-12 years), and adolescent (13-18 years). A comparison of the SUV values of each organ across age groups was performed. RESULTS The highest levels of physiological uptake were observed in the spleen across all age groups, except for infants, who demonstrated the highest SUV values in the kidneys. 68Ga-DOTA-TATE uptake in the parotid glands, submandibular glands, thyroid gland, thymus, liver, spleen, adrenal glands, stomach, intestines, uterus, prostate, and testes demonstrated a statistically significant increase in the adolescent age group. In contrast to all internal organs, the lowest SUV max values were observed for all growth plates within the adolescent age group. CONCLUSION This study presents the bio-distribution pattern of 68Ga-DOTA-TATE in pediatric patients, according to age and location. The ranges of the SUVmax and SUVmean values of 68Ga-DOTA-TATE obtained in the various organs are of paramount importance for accurately diagnosing malignancy in 68Ga-DOTA-TATE PET/CT studies.
Collapse
Affiliation(s)
- Nuh Filizoglu
- Department of Nuclear Medicine, University of Health Sciences, Kartal Dr. Lutfi Kirdar City Hospital, D-100 Güney Yanyol No:47 Cevizli Mevkii, Kartal, 34865, Istanbul, Turkey.
| | - Salih Ozguven
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Selin Kesim
- Department of Nuclear Medicine, University of Health Sciences, Kartal Dr. Lutfi Kirdar City Hospital, D-100 Güney Yanyol No:47 Cevizli Mevkii, Kartal, 34865, Istanbul, Turkey
| | - Kevser Oksuzoglu
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Feyza Caglıyan
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Tunc Ones
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Fuat Dede
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Halil Turgut Turoglu
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Tanju Yusuf Erdil
- Department of Nuclear Medicine, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Tsoy U, Pogosian K, Ryzhkova D, Yudina O, Yakovenko K, Ryazanov P, Matsueva I, Sokolnikova P, Salov M, Karonova T, Grineva E. Somatostatin Receptor Imaging in the Diagnosis and Management of Parathyroid Neuroendocrine Neoplasia. Diagnostics (Basel) 2024; 14:2718. [PMID: 39682626 PMCID: PMC11640021 DOI: 10.3390/diagnostics14232718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Parathyroid tumors are classified as parathyroid neuroendocrine neoplasia (NEN) by the IARC-WHO classification. These tumors can occur with NENs from other sites, which often require total-body [68Ga]-DOTA-peptides PET/CT. This study aimed to assess the utility of [68Ga]-DOTA-peptide PET/CT in imaging parathyroid NENs and to evaluate the underlying mechanisms. METHODS Fifty patients with primary hyperparathyroidism (PHPT) and parathyroid NENs histologically confirmed as parathyroid adenomas (PAs) were included. PET/CT with [68Ga]-DOTA-peptide was performed in 16 patients with localized PAs, including 10 with MEN1 syndrome. Somatostatin receptor types 2 and 5 (SST2 and SST5) staining was performed on PAs from 48 patients. Somatostatin analogs (SSA) were prescribed in four patients with MEN 1 syndrome and 1 with persistent acromegaly, all with PAs and PHPT. The therapy effects on calcium and parathyroid hormone (iPTH) were evaluated. RESULTS [68Ga]-DOTA-peptide PET/CT detected 20 PAs with high radiopharmaceutical uptake. SST2 expression was negative on PA cell membranes in all cases and positive on endothelium in 39 (81%) PAs. Membrane SST5 expression was positive in 25 (52%) PAs and endothelial was positive in 40 (83%). Serum calcium levels decreased in patients on SSA therapy, while iPTH did not. CONCLUSIONS PET/CT with [68Ga]-DOTA-peptides can detect parathyroid NENs. The incidental detection of high [68Ga]-DOTA-peptide uptake in the parathyroid region during whole-body PET/CT may prompt biochemical evaluation for PHPT. We suggest that endothelial SST expression mediates high radiopharmaceutical uptake by PAs and that SSA treatment can reduce hypercalcemia in PHPT patients.
Collapse
Affiliation(s)
| | - Karina Pogosian
- Almazov National Medical Research Centre, 2 Akkuratova Street, Saint Petersburg 197341, Russia; (U.T.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Hou G, Cheng X, Sun L, Wang X, Zheng R. A Rare Case of Follicular Dendritic Cell Tumor of the Abdominal Cavity Visualized by 18 F-AlF-NOTA-Octreotide PET/CT. Clin Nucl Med 2024; 49:1118-1119. [PMID: 38968592 DOI: 10.1097/rlu.0000000000005369] [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: 07/07/2024]
Abstract
ABSTRACT Positivity of follicular dendritic cell tumor in somatostatin receptor imaging is rare. A 68-year-old woman underwent ultrasound in health examination. The results showed abnormal echoes in the pancreatic head region, suggestive of a neuroendocrine tumor of the pancreatic head. 18 F-AlF-NOTA-octreotide PET/CT was performed for staging. 18 F-AlF-NOTA-octreotide PET/CT revealed an irregular mass between the caudate lobe of the liver and the pancreatic head with heterogeneously increased uptake, which was later confirmed as follicular dendritic cell tumor by pathological examination.
Collapse
Affiliation(s)
- Guozhu Hou
- From the Department of Nuclear Medicine (PET-CT Center)
| | - Xin Cheng
- From the Department of Nuclear Medicine (PET-CT Center)
| | - Li Sun
- Department of Pathology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuejuan Wang
- From the Department of Nuclear Medicine (PET-CT Center)
| | - Rong Zheng
- From the Department of Nuclear Medicine (PET-CT Center)
| |
Collapse
|
7
|
Varghese J, Skefos CM, Jimenez C. Metastatic pheochromocytoma and paraganglioma: Integrating tumor biology in clinical practice. Mol Cell Endocrinol 2024; 592:112344. [PMID: 39182716 DOI: 10.1016/j.mce.2024.112344] [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: 05/11/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumors derived from chromaffin cells in the autonomic nervous system. Depending on their location, these tumors are capable of excessive catecholamine production, which may lead to uncontrolled hypertension and other life-threatening complications. They are associated with a significant risk of metastatic disease and are often caused by an inherited germline mutation. Although surgery can cure localized disease and lead to remission, treatments for metastatic PPGL (mPPGL)-including chemotherapy, radiopharmaceutical agents, multikinase inhibitors, and immunotherapy used alone or in combination- aim to control tumor growth and limit organ damage. Substantial advances have been made in understanding hereditary and somatic molecular signaling pathways that play a role in tumor growth and metastasis. Treatment options for metastatic disease are rapidly evolving, and this paper aims to provide a brief overview of the management of mPPGL with a focus on therapy options.
Collapse
Affiliation(s)
- Jeena Varghese
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Catherine M Skefos
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Demeure MJ, Giuliano P, Forester J, Zakry N, Ulaner GA. 111 In-Pentetreotide-Guided Radiosurgery for Intraoperative Localization of Retroperitoneal Nodal Metastasis in a Patient With Ileal Neuroendocrine Tumor. Clin Nucl Med 2024; 49:695-697. [PMID: 38768160 DOI: 10.1097/rlu.0000000000005286] [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: 05/22/2024]
Abstract
ABSTRACT 64 Cu-DOTATATE PET/CT of a 44-year-old man with an ileal neuroendocrine tumor demonstrated the primary tumor, local nodal metastases, and a pericaval nodal metastasis. Localization of the pericaval node during surgery may be difficult, thus 4.4 mCi of 111 In-pentetreotide was administered before surgery to assist with localization and resection. At surgery, the pericaval nodal metastasis was readily detected by gamma probe, which could then be resected and pathologically proven to be a metastasis. This demonstrates the use of somatostatin receptor-targeted imaging for intraoperative localization of an otherwise difficult to surgically localize metastasis. Without intraoperative somatostatin receptor-targeted radiosurgery, disease may have been incompletely resected.
Collapse
|
9
|
Bertani E, Mattana F, Collamati F, Ferrari ME, Bagnardi V, Frassoni S, Pisa E, Mirabelli R, Morganti S, Fazio N, Fumagalli Romario U, Ceci F. Radio-Guided Surgery with a New-Generation β-Probe for Radiolabeled Somatostatin Analog, in Patients with Small Intestinal Neuroendocrine Tumors. Ann Surg Oncol 2024; 31:4189-4196. [PMID: 38652200 DOI: 10.1245/s10434-024-15277-x] [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: 01/11/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Radio-guided surgery (RGS) holds promise for improving surgical outcomes in neuroendocrine tumors (NETs). Previous studies showed low specificity (SP) using γ-probes to detect radiation emitted by radio-labeled somatostatin analogs. OBJECTIVE We aimed to assess the sensitivity (SE) and SP of the intraoperative RGS approach using a β-probe with a per-lesion analysis, while assessing safety and feasibility as secondary objectives. METHODS This prospective, single-arm, single-center, phase II trial (NCT05448157) enrolled 20 patients diagnosed with small intestine NETs (SI-NETs) with positive lesions detected at 68Ga-DOTA-TOC positron emission tomography/computed tomography (PET/CT). Patients received an intravenous injection of 1.1 MBq/Kg of 68Ga-DOTA-TOC 10 min prior to surgery. In vivo measurements were conducted using a β-probe. Receiver operating characteristic (ROC) analysis was performed, with the tumor-to-background ratio (TBR) as the independent variable and pathology result (cancer vs. non-cancer) as the dependent variable. The area under the curve (AUC), optimal TBR, and absorbed dose for the surgery staff were reported. RESULTS The intraoperative RGS approach was feasible in all cases without adverse effects. Of 134 specimens, the AUC was 0.928, with a TBR cut-off of 1.35 yielding 89.3% SE and 86.4% SP. The median absorbed dose for the surgery staff was 30 µSv (range 12-41 µSv). CONCLUSION This study reports optimal accuracy in detecting lesions of SI-NETs using the intraoperative RGS approach with a novel β-probe. The method was found to be safe, feasible, and easily reproducible in daily clinical practice, with minimal radiation exposure for the staff. RGS might potentially improve radical resection rates in SI-NETs. CLINICAL TRIALS REGISTRATION 68Ga-DOTATOC Radio-Guided Surgery with β-Probe in GEP-NET (RGS GEP-NET) [NCT0544815; https://classic. CLINICALTRIALS gov/ct2/show/NCT05448157 ].
Collapse
Affiliation(s)
- Emilio Bertani
- Neuroendocrine Surgery Tumor Unit, IEO, European Institute of Oncology IRCCS, Milan, Italy.
- Division of Digestive Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Francesco Mattana
- Division of Nuclear Medicine, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Mahila E Ferrari
- Division of Medical Physics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Samuele Frassoni
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Eleonora Pisa
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Riccardo Mirabelli
- Istituto Nazionale di Fisica Nucleare INFN, Sezione di Roma, Rome, Italy
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Rome, Italy
| | - Silvio Morganti
- Istituto Nazionale di Fisica Nucleare INFN, Sezione di Roma, Rome, Italy
| | - Nicola Fazio
- Division of Gastrointestinal and Neuroendocrine Tumors Medical Treatment IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Francesco Ceci
- Division of Nuclear Medicine, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
10
|
Edmonds S, Davidson J, Joseph R, Vyas M. Imaging Pathway of a Pediatric Patient with Succinate Dehydrogenase B-Deficient Paraganglioma. J Nucl Med Technol 2023; 51:318-322. [PMID: 37699643 DOI: 10.2967/jnmt.123.265655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 08/01/2023] [Indexed: 09/14/2023] Open
Abstract
Pheochromocytoma and paraganglioma are rare in children, at only 1 in every 50,000 cases. Even though some cases are sporadic, they have been connected to syndromes such as von Hippel-Lindau, multiple endocrine neoplasia types IIa and IIb, neurofibromatosis type 1, and hereditary pheochromocytoma-paraganglioma syndromes. A genetic mutation causes around 60% of pheochromocytomas and paragangliomas in children under 18. Methods: A 15-y-old child with a 6-y history of back discomfort is presented. The justification for using 2 functional imaging modalities, 68Ga-DOTATATE PET/CT and 123I-meta-iodobenzylguanidine SPECT/CT, is examined in this case study. We reviewed the patients' journey since the first referral for imaging. Results: Delaying the molecular imaging modalities has affected patients' overall diagnosis and applied treatment outcomes. Conclusion: This case study investigates the potential for the earlier use of various diagnostic modalities in conjunction with diagnostic testing to facilitate an earlier diagnosis. However, since this study is based solely on imaging and lacks access to the patient's clinical or family history, factors such as potential inequities in health-care facilities, health literacy, and socioeconomic status are not addressed. It is essential to acknowledge these influences as they contribute to the inequitable access to health-care settings in New Zealand.
Collapse
Affiliation(s)
| | - Jennifer Davidson
- Mercy Radiology, Auckland, New Zealand
- Auckland Hospital, Auckland, New Zealand; and
| | | | - Madhusudan Vyas
- Mercy Radiology, Auckland, New Zealand;
- Unitec Institute of Technology, Auckland, New Zealand
| |
Collapse
|
11
|
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
|
12
|
Qi M, Liu Y, Su M, Huang R. Primary Hepatocellular Carcinoma Revealed on 68Ga-DOTATATE in a Patient With Nasopharyngeal Carcinoma. Clin Nucl Med 2023:00003072-990000000-00642. [PMID: 37486702 DOI: 10.1097/rlu.0000000000004774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
ABSTRACT A 56-year-old man underwent a prospective study (ChiCTR2300070081), which is a head-to-head comparison of 18F-FDG and 68Ga-DOTATATE PET/MR in EB-positive nonkeratinizing nasopharyngeal carcinoma after chemotherapy. Bilateral cervical abnormal lymph nodes were both detected by 18F-FDG and 68Ga-DOTATATE PET/MRI, whereas 2 hepatic lesions only were shown on 68Ga-DOTATATE, which subsequent pathologically proved to be primary hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mengfang Qi
- From the Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, China
| | | | | | | |
Collapse
|
13
|
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: 8] [Impact Index Per Article: 4.0] [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
|
14
|
Bocchini M, Tazzari M, Ravaioli S, Piccinini F, Foca F, Tebaldi M, Nicolini F, Grassi I, Severi S, Calogero RA, Arigoni M, Schrader J, Mazza M, Paganelli G. Circulating hsa-miR-5096 predicts 18F-FDG PET/CT positivity and modulates somatostatin receptor 2 expression: a novel miR-based assay for pancreatic neuroendocrine tumors. Front Oncol 2023; 13:1136331. [PMID: 37287922 PMCID: PMC10242108 DOI: 10.3389/fonc.2023.1136331] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/25/2023] [Indexed: 06/09/2023] Open
Abstract
Gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs) are rare diseases encompassing pancreatic (PanNETs) and ileal NETs (SINETs), characterized by heterogeneous somatostatin receptors (SSTRs) expression. Treatments for inoperable GEP-NETs are limited, and SSTR-targeted Peptide Receptor Radionuclide Therapy (PRRT) achieves variable responses. Prognostic biomarkers for the management of GEP-NET patients are required. 18F-FDG uptake is a prognostic indicator of aggressiveness in GEP-NETs. This study aims to identify circulating and measurable prognostic miRNAs associated with 18F-FDG-PET/CT status, higher risk and lower response to PRRT. Methods Whole miRNOme NGS profiling was conducted on plasma samples obtained from well-differentiated advanced, metastatic, inoperable G1, G2 and G3 GEP-NET patients enrolled in the non-randomized LUX (NCT02736500) and LUNET (NCT02489604) clinical trials prior to PRRT (screening set, n= 24). Differential expression analysis was performed between 18F-FDG positive (n=12) and negative (n=12) patients. Validation was conducted by Real Time quantitative PCR in two distinct well-differentiated GEP-NET validation cohorts, considering the primary site of origin (PanNETs n=38 and SINETs n=30). The Cox regression was applied to assess independent clinical parameters and imaging for progression-free survival (PFS) in PanNETs. In situ RNA hybridization combined with immunohistochemistry was performed to simultaneously detect miR and protein expression in the same tissue specimens. This novel semi-automated miR-protein protocol was applied in PanNET FFPE specimens (n=9). In vitro functional experiments were performed in PanNET models. Results While no miRNAs emerged to be deregulated in SINETs, hsa-miR-5096, hsa-let-7i-3p and hsa-miR-4311 were found to correlate with 18F-FDG-PET/CT in PanNETs (p-value:<0.005). Statistical analysis has shown that, hsa-miR-5096 can predict 6-month PFS (p-value:<0.001) and 12-month Overall Survival upon PRRT treatment (p-value:<0.05), as well as identify 18F-FDG-PET/CT positive PanNETs with worse prognosis after PRRT (p-value:<0.005). In addition, hsa-miR-5096 inversely correlated with both SSTR2 expression in PanNET tissue and with the 68Gallium-DOTATOC captation values (p-value:<0.05), and accordingly it was able to decrease SSTR2 when ectopically expressed in PanNET cells (p-value:<0.01). Conclusions hsa-miR-5096 well performs as a biomarker for 18F-FDG-PET/CT and as independent predictor of PFS. Moreover, exosome-mediated delivery of hsa-miR-5096 may promote SSTR2 heterogeneity and thus resistance to PRRT.
Collapse
Affiliation(s)
- Martine Bocchini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Marcella Tazzari
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Ravaioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Filippo Piccinini
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Michela Tebaldi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Ilaria Grassi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Raffaele Adolfo Calogero
- Molecular Biotechnology Center, Department of Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Maddalena Arigoni
- Molecular Biotechnology Center, Department of Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Joerg Schrader
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Massimiliano Mazza
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
15
|
PET Criteria by Cancer Type from Imaging Interpretation to Treatment Response Assessment: Beyond FDG PET Score. Life (Basel) 2023; 13:life13030611. [PMID: 36983767 PMCID: PMC10057339 DOI: 10.3390/life13030611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 01/30/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Background: in recent years, the role of positron emission tomography (PET) and PET/computed tomography (PET/CT) has emerged as a reliable diagnostic tool in a wide variety of pathological conditions. This review aims to collect and review PET criteria developed for interpretation and treatment response assessment in cases of non-[18F]fluorodeoxyglucose ([18F]FDG) imaging in oncology. Methods: A wide literature search of the PubMed/MEDLINE, Scopus and Google Scholar databases was made to find relevant published articles about non-[18F]FDG PET response criteria. Results: The comprehensive computer literature search revealed 183 articles. On reviewing the titles and abstracts, 149 articles were excluded because the reported data were not within the field of interest. Finally, 34 articles were selected and retrieved in full-text versions. Conclusions: available criteria are a promising tool for the interpretation of non-FDG PET scans, but also to assess the response to therapy and therefore to predict the prognosis. However, oriented clinical trials are needed to clearly evaluate their impact on patient management.
Collapse
|
16
|
Si Z, Cheng Y, Xu Z, Shi D, Shi H, Cheng D. Exploration of 68Ga-DOTA-MAL as a Versatile Vehicle for Facile Labeling of a Variety of Thiol-Containing Bioactive Molecules. ACS OMEGA 2023; 8:4747-4755. [PMID: 36777559 PMCID: PMC9909812 DOI: 10.1021/acsomega.2c06720] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Efficient and site-specific radiolabeling reactions are essential in molecular probe synthesis. Thus, selecting an effective method for radiolabeling that does not affect bioactivity of the molecule is critical. Varieties of bifunctional chelating agents provide a solution in this matter. As a chemo-specific chelator, maleimido-mono-amide-DOTA (DOTA-Mal) holds significant potential for 68Ga labeling of bioactive molecules; it can react specifically with free sulfhydryl groups under mild conditions. Compared with amino and carboxylic acid groups, free sulfhydryl groups are relatively less common in most biomolecules and can serve as site-specific radiolabeling targets. Labeling of 68Ga usually employs a two-step labeling strategy; first, chelators are conjugated to the biomolecules, which is followed by radiolabeling. However, the bioactivity of biomolecules may be affected by harsh labeling conditions. In this study, three 68Ga-labeled bioactive molecules, namely, 68Ga-DOTA-RGD, 68Ga-DOTA-FA, and 68Ga-DOTA-BSA, were prepared using a novel strategy under mild conditions (pH of 8.0 at room temperature). Using this strategy, DOTA-Mal was labeled by 68Ga before it reacted with the sulfhydryl group-containing biomolecules, which avoided damage to said biomolecules caused by the harsh reaction conditions required in 68Ga-labeling procedures. The biological and chemical properties of these three radiotracers synthesized using this strategy are well manifested. Through a series of experiments, the effectiveness of this strategy is demonstrated, and we believe that this site-specific bioactivity-friendly reaction strategy will facilitate developments and translation applications of varieties of 68Ga-labeled positron emission tomography probes.
Collapse
Affiliation(s)
- Zhan Si
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| | - Yuan Cheng
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| | - Zhan Xu
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| | - Dai Shi
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| | - Hongcheng Shi
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| | - Dengfeng Cheng
- Department
of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Institute
of Nuclear Medicine, Fudan University, Shanghai 200032, China
- Shanghai
Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
17
|
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
|
18
|
Zahed H, Beauregard JM, Abikhzer G, Rush C, Probst S. Differential Detection of Hepatic Metastases on 68 Ga-DOTATATE PET/CT and 177 Lu-DOTATATE SPECT/CT. Clin Nucl Med 2023; 48:e12-e15. [PMID: 36240803 DOI: 10.1097/rlu.0000000000004434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT 68 Ga-DOTATATE PET/CT is indicated for selecting patients for peptide receptor radionuclide therapy (PRRT). Although highly sensitive, the detectability of smaller lesions, particularly in the liver, is lower. We present the case of a 58-year-old man with metastatic well-differentiated pancreatic neuroendocrine tumor whose MRI revealed progression of hepatic metastases. 68 Ga-DOTATATE PET/CT performed to determine eligibility for PRRT did not demonstrate DOTATATE-avid disease within the liver. 18 F-FDG PET/CT was also negative at the liver and the patient proceeded to 177 Lu-DOTATATE PRRT, where multi-time point posttherapy planar imaging and SPECT/CT showed intense uptake in the known liver metastases.
Collapse
Affiliation(s)
- Hanan Zahed
- From the Department of Nuclear Medicine, Jewish General Hospital, McGill University, Montreal
| | - Jean-Mathieu Beauregard
- Department of Medical Imaging, CHU de Québec-Université Laval; and Department of Radiology and Nuclear Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Gad Abikhzer
- From the Department of Nuclear Medicine, Jewish General Hospital, McGill University, Montreal
| | - Christopher Rush
- From the Department of Nuclear Medicine, Jewish General Hospital, McGill University, Montreal
| | - Stephan Probst
- From the Department of Nuclear Medicine, Jewish General Hospital, McGill University, Montreal
| |
Collapse
|
19
|
Stenvall A, Gustafsson J, Larsson E, Roth D, Sundlöv A, Jönsson L, Hindorf C, Ohlsson T, Sjögreen Gleisner K. Relationships between uptake of [ 68Ga]Ga-DOTA-TATE and absorbed dose in [ 177Lu]Lu-DOTA-TATE therapy. EJNMMI Res 2022; 12:75. [PMID: 36534192 PMCID: PMC9763525 DOI: 10.1186/s13550-022-00947-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Somatostatin receptor 68Ga PET imaging is standard for evaluation of a patient's suitability for 177Lu peptide receptor radionuclide therapy of neuroendocrine tumours (NETs). The 68Ga PET serves to ensure sufficient somatostatin receptor expression, commonly evaluated qualitatively. The aim of this study is to investigate the quantitative relationships between uptake in 68Ga PET and absorbed doses in 177Lu therapy. METHOD Eighteen patients underwent [68Ga]Ga-DOTA-TATE PET imaging within 20 weeks prior to their first cycle of [177Lu]Lu-DOTA-TATE. Absorbed doses for therapy were estimated for tumours, kidney, spleen, and normal liver parenchyma using a hybrid SPECT/CT-planar method. Gallium-68 activity concentrations were retrieved from PET images and also used to calculate SUVs and normalized SUVs, using blood and tissue for normalization. The 68Ga activity concentrations per injected activity, SUVs, and normalized SUVs were compared with 177Lu activity concentrations 1 d post-injection and 177Lu absorbed doses. For tumours, for which there was a variable number per patient, both inter- and intra-patient correlations were analysed. Furthermore, the prediction of 177Lu tumour absorbed doses based on a combination of tumour-specific 68Ga activity concentrations and group-based estimates of the effective half-lives for grade 1 and 2 NETs was explored. RESULTS For normal organs, only spleen showed a significant correlation between the 68Ga activity concentration and 177Lu absorbed dose (r = 0.6). For tumours, significant, but moderate, correlations were obtained, with respect to both inter-patient (r = 0.7) and intra-patient (r = 0.45) analyses. The correlations to absorbed doses did not improve when using 68Ga SUVs or normalized SUVs. The relationship between activity uptakes for 68Ga PET and 177Lu SPECT was stronger, with correlation coefficients r = 0.8 for both inter- and intra-patient analyses. The 177Lu absorbed dose to tumour could be predicted from the 68Ga activity concentrations with a 95% coverage interval of - 65% to 248%. CONCLUSIONS On a group level, a high uptake of [68Ga]Ga-DOTA-TATE is associated with high absorbed doses at 177Lu-DOTA-TATE therapy, but the relationship has a limited potential with respect to individual absorbed dose planning. Using SUV or SUV normalized to reference tissues do not improve correlations compared with using activity concentration per injected activity.
Collapse
Affiliation(s)
- Anna Stenvall
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Johan Gustafsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Erik Larsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Daniel Roth
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden
| | - Anna Sundlöv
- grid.4514.40000 0001 0930 2361Division of Oncology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Lena Jönsson
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Cecilia Hindorf
- grid.4514.40000 0001 0930 2361Medical Radiation Physics, Lund, Lund University, Lund, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Ohlsson
- grid.411843.b0000 0004 0623 9987Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
20
|
Gastric neuroendocrine neoplasms: a primer for radiologists. ABDOMINAL RADIOLOGY (NEW YORK) 2022; 47:3993-4004. [PMID: 35411433 DOI: 10.1007/s00261-022-03509-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 01/18/2023]
Abstract
Gastric neuroendocrine neoplasms are uncommon tumors with variable differentiation and malignant potential. Three main subtypes are recognized: type 1, related to autoimmune atrophic gastritis; type 2, associated with Zollinger-Ellison and MEN1 syndrome; and type 3, sporadic. Although endoscopy alone is often sufficient for diagnosis and management of small, indolent, multifocal type 1 tumors, imaging is essential for evaluation of larger, high-grade, and type 2 and 3 neoplasms. Hypervascular intraluminal gastric masses are typically seen on CT/MRI, with associated perigastric lymphadenopathy and liver metastases in advanced cases. Somatostatin receptor nuclear imaging (such as Ga-68-DOTATATE PET/CT) may also be used for staging and assessing candidacy for peptide receptor radionuclide therapy. Radiotracer uptake is more likely in well-differentiated, lower-grade tumors, and less likely in poorly differentiated tumors, for which F-18-FDG-PET/CT may have additional value. Understanding disease pathophysiology and evolving histologic classifications is particularly useful for radiologists, as these influence tumor behavior, preferred imaging, therapy options, and patient prognosis.
Collapse
|
21
|
Small bowel neuroendocrine neoplasm: what surgeons want to know. ABDOMINAL RADIOLOGY (NEW YORK) 2022; 47:4005-4015. [PMID: 35312820 DOI: 10.1007/s00261-022-03485-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 01/18/2023]
Abstract
Neuroendocrine neoplasms of the small bowel are a diverse group of tumors with a broad spectrum of imaging findings and clinical implications. Most tumors originate in close proximity to the ileocecal valve and most commonly metastasize to the mesentery and liver. This review will highlight the imaging findings of primary and metastatic small bowel neuroendocrine neoplasm that are most relevant to the surgical team.
Collapse
|
22
|
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
|
23
|
Takagi H, Matsumura Y, Fukuhara M, Inomata S, Yamaguchi H, Watanabe M, Ozaki Y, Muto S, Okabe N, Shio Y, Saito H, Tanabe H, Shimabukuro M, Suzuki H. Ectopic adrenocorticotropic hormone-secreting carcinoid with solitary cryptococcosis in the lungs. Cancer Rep (Hoboken) 2022; 5:e1731. [PMID: 36196010 PMCID: PMC9675365 DOI: 10.1002/cnr2.1731] [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: 10/18/2021] [Revised: 08/30/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Carcinoid tumors can on rare occasions ectopically produce adrenocorticotropic hormone (ACTH), causing Cushing's syndrome, and patients could become immunocompromised. Care must therefore be taken regarding infectious complications. In particular, ACTH-producing pulmonary carcinoid is not easy to diagnose by itself, and when combined with pulmonary nodules as infectious foci, each is very difficult to diagnose. CASE The patient was a 71-year-old woman with refractory diabetes. She showed clinical symptoms of Cushing's syndrome during treatment for diabetes and ectopic ACTH production was suspected based on biochemical and imaging tests. Nodules were identified in the left lung apex and lingual segment. Examination of resected nodules revealed that the nodule in the apex was pulmonary cryptococcosis, while the nodule in the lingual segment represented typical carcinoid. After surgery, clinical symptoms, laboratory findings, and diabetes all improved. CONCLUSION We present this very instructive case in terms of the difficulty of diagnosing ACTH-producing tumors, the possibility of infection complicating the immunodeficiency caused by ACTH-producing tumors, and the surgical strategy.
Collapse
Affiliation(s)
- Hironori Takagi
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan,Department of Chest SurgeryIwaki City Medical CenterIwakiJapan
| | - Yuki Matsumura
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | | | - Sho Inomata
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Hikaru Yamaguchi
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Masayuki Watanabe
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Yuki Ozaki
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Satoshi Muto
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Naoyuki Okabe
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Yutaka Shio
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| | - Haruka Saito
- Department of Diabetes, Endocrinology and MetabolismFukushima Medical UniversityFukushimaJapan
| | - Hayato Tanabe
- Department of Diabetes, Endocrinology and MetabolismFukushima Medical UniversityFukushimaJapan
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and MetabolismFukushima Medical UniversityFukushimaJapan
| | - Hiroyuki Suzuki
- Department of Chest SurgeryFukushima Medical UniversityFukushimaJapan
| |
Collapse
|
24
|
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: 8] [Impact Index Per Article: 2.7] [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
|
25
|
Engur CO, Apaydın T, Ones T, Gozu HI, Ozguven S. Corticomedullary Mixed Tumor of the Adrenal Gland with Apparent 18F-Fluorodeoxyglucose Activity But No 68GA-DOTATATE Uptake on Positron Emission Tomography/Computed Tomography. Indian J Nucl Med 2022; 37:297-298. [PMID: 36686306 PMCID: PMC9855236 DOI: 10.4103/ijnm.ijnm_194_21] [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: 12/06/2021] [Revised: 02/27/2022] [Accepted: 03/16/2022] [Indexed: 01/24/2023] Open
Abstract
Corticomedullary mixed tumor (CMT) is a single adrenal tumor mass composed histologically by an admixture of adrenal cortical and medullary cells. It is a rare condition, with approximately 20 cases reported to date. To our knowledge, the positron emission tomography (PET) imaging findings of this mostly benign tumor have not been reported in the literature. We present a case of CMT who was evaluated with both 18F-fluorodeoxyglucose (18F-FDG) and 68Ga-DOTATATE. The hypermetabolic tumor seen on 18F-FDG PET/computed tomography scan showed no abnormal uptake by 68Ga-DOTATATE.
Collapse
Affiliation(s)
- Ceren Ozge Engur
- Department of Nuclear Medicine, Marmara University, Pendik Training and Research Hospital, Istanbul, Turkey
| | - Tugce Apaydın
- Department of Endocrinology and Metabolism, Marmara University, Pendik Training and Research Hospital, Istanbul, Turkey
| | - Tunc Ones
- Department of Nuclear Medicine, Marmara University, Pendik Training and Research Hospital, Istanbul, Turkey
| | - Hulya Iliksu Gozu
- Department of Endocrinology and Metabolism, Marmara University, Pendik Training and Research Hospital, Istanbul, Turkey
| | - Salih Ozguven
- Department of Nuclear Medicine, Marmara University, Pendik Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
26
|
Soulek DK, Mastascusa NJ, Martin ME, Graves SA. Practical Considerations for Implementation of 177Lu-DOTATATE Neuroendocrine Tumor Treatment Programs. J Nucl Med Technol 2022; 50:jnmt.122.263813. [PMID: 35701215 DOI: 10.2967/jnmt.122.263813] [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: 01/10/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
The 2018 FDA approval of 177Lu-DOTATATE for the treatment of somatostatin receptor-positive (SSTR) neuroendocrine tumors (NETs) represents a paradigm shifting approach to cancer treatments around the globe. Gastroenteropancreatic (GEP) NETs overexpress the somatostatin subtype receptor 2, which is now exploited for receptor-based imaging and therapy, thus generating significant progress in the diagnosis and treatment of this orphan disease. The recent FDA approval of receptor-based PET radiopharmaceuticals and a new peptide receptor radiopharmaceutical therapy (PRRT), 177Lu-DOTATATE, has dramatically impacted NET patient management. The focus of this paper is to review clinical considerations associated with implementing a 177Lu-DOTATATE program. We review receptor-based NET radiopharmaceuticals, 177Lu-DOTATATE patient selection criteria, administration methods, clinical, regulatory, and radiation safety considerations, technical factors, tissue dosimetry, and reimbursement guidelines.
Collapse
|
27
|
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
|
28
|
Grey N, Silosky M, Lieu CH, Chin BB. Current status and future of targeted peptide receptor radionuclide positron emission tomography imaging and therapy of gastroenteropancreatic-neuroendocrine tumors. World J Gastroenterol 2022; 28:1768-1780. [PMID: 35633909 PMCID: PMC9099199 DOI: 10.3748/wjg.v28.i17.1768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/07/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Theranostics is the highly targeted molecular imaging and therapy of tumors. Targeted peptide receptor radionuclide therapy has taken the lead in demonstrating the safety and effectiveness of this molecular approach to treating cancers. Metastatic, well-differentiated gastroenteropancreatic neuroendocrine tumors may be most effectively imaged and treated with DOTATATE ligands. We review the current practice, safety, advantages, and limitations of DOTATATE based theranostics. Finally, we briefly describe the exciting new areas of development and future directions of gastroenteropancreatic neuroendocrine tumor theranostics.
Collapse
Affiliation(s)
- Neil Grey
- Radiology-Nuclear Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Michael Silosky
- Department of Radiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Christopher H Lieu
- Medical Oncology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Bennett B Chin
- Department of Radiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
29
|
McClenathan M, Peacock JG. The "Ultimate Triple PET": 68Ga-DOTATATE, 18F-FDG, and 18F-Fluciclovine PET/CT Findings in a Single Patient With Metastatic Abdominal Carcinoid and Prostate Cancer. Clin Nucl Med 2022; 47:e161-e164. [PMID: 34392283 DOI: 10.1097/rlu.0000000000003853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT A 71-year-old man with a history of carcinoid and prostate adenocarcinoma underwent 68Ga-DOTATATE PET/CT to evaluate new mediastinal lymphadenopathy. The scan revealed new carcinoid metastases, but the mediastinal nodes were not avid. 18F-FDG PET/CT was performed to evaluate for cardiac and mediastinal sarcoidosis, revealing a lack of cardiac sarcoidosis, but probable mediastinal sarcoidosis. 18F-Fluciclovine PET/CT was then performed for possible prostate metastases, revealing the patient's known prostate cancer. Three different PET radiopharmaceuticals revealed 3 different processes in a single patient within a 195-day period, demonstrating the power of nuclear medicine diagnostics.
Collapse
Affiliation(s)
- Matthew McClenathan
- From the Department of Radiology, Brooke Army Medical Center, San Antonio, TX
| | | |
Collapse
|
30
|
Ragab A, Wu J, Ding X, Clark A, Mischen B, Chauhan A, Oates ME, Anthony L, El Khouli R. 68Ga-DOTATATE PET/CT: The Optimum Standardized Uptake Value (SUV) Internal Reference. Acad Radiol 2022; 29:95-106. [PMID: 34756348 DOI: 10.1016/j.acra.2020.08.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023]
Abstract
RATIONALE AND OBJECTIVES Standardized Uptake Value (SUV) is an important semiquantitative measurement used in the clinical and research domains to assess radiopharmaceutical concentration in tumors versus normal organs, but is susceptible to many factors beyond the tumor biological environment. So, the aim of this study is to identify the optimum internal reference among organs with physiological uptake in 68Ga-DOTATATE PET/CT (DOTA PET/CT) scans. MATERIALS AND METHODS This HIPAA-compliant, IRB-approved study with waiver of consent included retrospective imaging review of 180 consecutive patients with neuroendocrine tumors presenting for DOTA PET/CT image acquisition: Ga-68 DOTATATE dose was reported as (0.054 mCi/Kg) scans between September 2018 and May 2019. Mean value of body weight normalized SUV (SUVbw) and lean body mass normalized SUV (SUL) of liver and spleen were measured. Information about the patients and scan characteristics were collected. The paired Grambsch test was used to compare variance among the measured SUVs. Spearman's rank correlation coefficient was used to assess correlation between SUVs and potential patient- and scan-specific confounding factors. RESULTS Variance of SUL was significantly lower than variance of SUVbw in both liver and spleen (p-value < 0.0001). Variances of liver SUVbw and SUL were significantly lower than the corresponding spleen SUVs. Liver SUL showed the lowest variance (3.69% ± 1.25%) among all measured SUVs. CONCLUSION SUL is a more reproducible, less variable, and therefore more reliable quantitative measure in DOTA PET/CT scans, compared SUVbw. Among the available organs with physiological uptake, liver SUL is the optimum internal reference given the liver's larger size and uniform SUL values resulting in lower variability and better reproducibility.
Collapse
Affiliation(s)
- Ahmed Ragab
- Yale New Haven Health - Bridgeport Hospital, Bridgeport, Connecticut
| | - Jianrong Wu
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Cancer Biostatistics, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Biostatistics and Bioinformatics Shared Resource Facility, Lexington, Kentucky
| | - Xue Ding
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Cancer Biostatistics, Lexington, Kentucky
| | - Aurela Clark
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Blaine Mischen
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Aman Chauhan
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Medical Oncology, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky
| | - M Elizabeth Oates
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY
| | - Lowell Anthony
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Medical Oncology, Lexington, Kentucky; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky
| | - Riham El Khouli
- University of Kentucky College of Medicine, Department of Radiology, Division of Nuclear Medicine and Molecular imaging, 800 Rose street, Lexington, 40536 KY; University of Kentucky College of Medicine, Markey Cancer Center, Lexington, Kentucky.
| |
Collapse
|
31
|
Jayaprakasam VS, Paroder V, Schöder H. Variants and Pitfalls in PET/CT Imaging of Gastrointestinal Cancers. Semin Nucl Med 2021; 51:485-501. [PMID: 33965198 PMCID: PMC8338802 DOI: 10.1053/j.semnuclmed.2021.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past two decades, PET/CT has become an essential modality in oncology increasingly used in the management of gastrointestinal (GI) cancers. Most PET/CT tracers used in clinical practice show some degree of GI uptake. This uptake is quite variable and knowledge of common patterns of biodistribution of various radiotracers is helpful in clinical practice. 18F-Fluoro-Deoxy-Glucose (FDG) is the most commonly used radiotracer and has quite a variable uptake within the bowel. 68Ga-Prostate specific membrane antigen (PSMA) shows intense uptake within the proximal small bowel loops. 11C-methyl-L-methionine (MET) shows high accumulation within the bowels, which makes it difficult to assess bowel or pelvic diseases. One must also be aware of technical artifacts causing difficulties in interpretations, such as high attenuation oral contrast material within the bowel lumen or misregistration artifact due to patient movements. It is imperative to know the common variants and benign diseases that can mimic malignant pathologies. Intense FDG uptake within the esophagus and stomach may be a normal variant or may be associated with benign conditions such as esophagitis, reflux disease, or gastritis. Metformin can cause diffuse intense uptake throughout the bowel loops. Intense physiologic uptake can also be seen within the anal canal. Segmental bowel uptake can be seen in inflammatory bowel disease, radiation, or medication induced enteritis/colitis or infection. Diagnosis of appendicitis or diverticular disease requires CT correlation, as normal appendix or diverticulum can show intense uptake. Certain malignant pathologies are known to have only low FDG uptake, such as early-stage esophageal adenocarcinoma, mucinous tumors, indolent lymphomas, and multicystic mesotheliomas. Response assessment, particularly in the neoadjuvant setting, can be limited by post-treatment inflammatory changes. Post-operative complications such as abscess or fistula formation can also show intense uptake and may obscure underlying malignant pathology. In the absence of clinical suspicion or rising tumor marker, the role of FDG PET/CT in routine surveillance of patients with GI malignancy is not clear.
Collapse
Affiliation(s)
- Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viktoriya Paroder
- Body Imaging Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heiko Schöder
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
32
|
Siebinga H, de Wit-van der Veen BJ, Beijnen JH, Stokkel MPM, Dorlo TPC, Huitema ADR, Hendrikx JJMA. A physiologically based pharmacokinetic (PBPK) model to describe organ distribution of 68Ga-DOTATATE in patients without neuroendocrine tumors. EJNMMI Res 2021; 11:73. [PMID: 34398356 PMCID: PMC8368277 DOI: 10.1186/s13550-021-00821-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background Physiologically based pharmacokinetic (PBPK) models combine drug-specific information with prior knowledge on the physiology and biology at the organism level. Whole-body PBPK models contain an explicit representation of the organs and tissue and are a tool to predict pharmacokinetic behavior of drugs. The aim of this study was to develop a PBPK model to describe organ distribution of 68Ga-DOTATATE in a population of patients without detectable neuroendocrine tumors (NETs). Methods Clinical 68Ga-DOTATATE PET/CT data from 41 patients without any detectable somatostatin receptor (SSTR) overexpressing tumors were included. Scans were performed at 45 min (range 30–60 min) after intravenous bolus injection of 68Ga-DOTATATE. Organ (spleen, liver, thyroid) and blood activity levels were derived from PET scans, and corresponding DOTATATE concentrations were calculated. A whole-body PBPK model was developed, including an internalization reaction, receptor recycling, enzymatic reaction for intracellular degradation and renal clearance. SSTR2 expression was added for several organs. Input parameters were fixed or estimated using a built-in Monte Carlo algorithm for parameter identification. Results 68Ga-DOTATATE was administered with a median peptide amount of 12.3 µg (range 8.05–16.9 µg) labeled with 92.7 MBq (range 43.4–129.9 MBq). SSTR2 amounts for spleen, liver and thyroid were estimated at 4.40, 7.80 and 0.0108 nmol, respectively. Variability in observed organ concentrations was best described by variability in SSTR2 expression and differences in administered peptide amounts. Conclusions To conclude, biodistribution of 68Ga-DOTATATE was described with a whole-body PBPK model, where tissue distribution was mainly determined by variability in SSTR2 organ expression and differences in administered peptide amounts.
Collapse
Affiliation(s)
- H Siebinga
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - B J de Wit-van der Veen
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M P M Stokkel
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T P C Dorlo
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands. .,Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Abstract
Consensus guidelines acknowledge the role of gallium Ga-68 (68Ga) 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic (DOTA) somatostatin receptor (SSTR) positron emission tomography/computed tomography (PET/CT) in management of neuroendocrine tumor (NET) patients. 68Ga-DOTA-SSTR PET/CT demonstrates superior performance to conventional imaging in initial detection, staging, detection of recurrent tumor, and detection of unknown primary in known metastatic disease. 68Ga-DOTA-SSTR PET/CT is low yield for NET detection in the setting of symptoms or elevated biomarkers when conventional imaging is negative, but may still guide management. The role of 68Ga-DOTA-SSTR PET/CT is not established in monitoring response to systemic therapy but may identify progression through detection of new metastases.
Collapse
Affiliation(s)
- Janet Pollard
- Department of Radiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| | - Parren McNeely
- Department of Radiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Yusuf Menda
- Department of Radiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| |
Collapse
|
34
|
Standardized uptake values and ratios on 68Ga-DOTATATE PET-computed tomography for normal organs and malignant lesions and their correlation with Krenning score in patients with metastatic neuroendocrine tumors. Nucl Med Commun 2021; 41:1095-1099. [PMID: 32732598 DOI: 10.1097/mnm.0000000000001253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim was to estimate the physiological standardized uptake values (SUVs) on Ga-DOTATATE PET-computed tomography (CT) in normal organs and metastatic tumor lesions (both standard and delayed), and correlating the uptake values and ratios with Krenning Scores (K-score) in patients with metastatic/advanced neuroendocrine tumors (NETs) undergoing PET-CT studies for their management work-up. A total of 32 patients of metastatic NET with 95 discrete tumor lesions were included in this analysis. These patients underwent standard whole-body PET-CT following injection of 2-3 mCi (74-111 MBq) of Ga-DOTATATE at 1-1.5 h. The normal physiological SUVmean of the liver and spleen and SUVmax and SUVmean of tumor lesions were estimated by an in-built automated procedure. These patients also underwent a delayed scan (2.5-3 h) and the same parameters were obtained for the delayed study. The tumorous lesions were further classified on the basis of K-score, and this was correlated with the mean SUVmax on both early and delayed scans. SUVmean ratios (tumor-to-liver and tumor-to-spleen) were also calculated for both time-points and correlated with individual K-scores. In lesions with K-score 4, the mean SUVmax was 32.5 in early and 30.5 in delayed scan, for lesions with K-score of 3 and 2, the mean SUVmax were 17.3, 20, and 9.3, 9.2, respectively, while in K-score 1 (n = 1), the delayed mean SUVmax was found to be more than early mean SUVmax (3.2 to 2.3). Statistical significance was evaluated by paired t test, and the changes in SUVmax was found to be statistically insignificant (P > 0.05) in all 3 K-scores. The paired t test was also performed between early and delayed tumor/liver and tumor/spleen mean SUVmean ratios, and no significant changes were observed across all K scores. The mean SUVmean values of the liver in the standard 1-h scan and delayed scans were 8.05 (range: 3-15) and 8.17 (range: 3.2-16), while for spleen, the values were 18 (range: 8.4-36.7) and 20 (range: 10-38.6), respectively. Statistically significant changes were observed in delayed spleen SUVmean values compared to the early scan (P < 0.05), while for liver SUVmean, the difference was not significant. Thus, in the present study, the SUVmax and SUVmean (range and mean values) for normal liver and spleen, and malignant NET lesions, and tumor-to-liver and tumor-to-spleen SUVmean ratios of different K-scores were generated. As could be theoretically expected in receptor-based PET-CT, there was no significant change in the delayed scan compared to the standard 1-1.5 h values.
Collapse
|
35
|
Ga-68 DOTATATE PET/CT and F-18 FDG PET/CT in the evaluation of low and intermediate versus high-grade neuroendocrine tumors. Nucl Med Commun 2021; 41:1060-1065. [PMID: 32732600 DOI: 10.1097/mnm.0000000000001255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We investigated the role of Ga-68 DOTATATE PET/CT in comparison to F-18 FDG PET/CT in patients with low and intermediate versus high-grade neuroendocrine tumors (NETs). METHODS We identified 81 patients who underwent Ga-68 DOTATATE PET/CT at our institution between May 2017 and December 2018 and met inclusion criteria of biopsy-proven NET with known Ki-67 index, histologic grade, or differentiation. Patients were divided into two groups. Control group included Ki-67 ≤20%, grade 1 or 2, or well-differentiated tumors. Experimental group included Ki-67 >20%, grade 3, or poorly-differentiated tumors. RESULTS Mean age was 57 years, with 36 males and 45 females. Most common primary sites were small bowel, pancreas, and lung. Most common distant metastatic sites were liver and bone. In the control group (n = 67), median Ki-67 was 4% (range 1-30%). 55/67 (82.1%) DOTATATE and 6/11 (54.5%) FDG scans were positive (P = 0.04). Positive scans showed >10 lesions in 25/55 (45.5%) DOTATATE and 1/6 (16.7%) FDG scans (P = 0.18). 40/55 (72.7%) positive DOTATATE and 3/6 (50%) FDG scans showed distant disease (P = 0.25). In the experimental group (n = 14), median Ki-67 was 68% (range 25-95%). All 14 DOTATATE and all nine FDG scans were positive. Positive scans showed >10 lesions in 4/14 (28.6%) DOTATATE and 5/9 (55.6%) FDG scans (P = 0.20). 10/14 (71.4%) positive DOTATATE and 7/9 (77.8%) FDG scans showed distant disease (P = 0.74). CONCLUSION All patients with high grade, poorly-differentiated NETs had positive DOTATATE PET/CTs. In these patients, DOTATATE PET/CT did not significantly differ from FDG PET/CT in identifying >10 lesions or distant disease.
Collapse
|
36
|
Moffat D, Richards P, Kurzawinski TR, Khan S, Khoo B, Grossman A. Misleading 68 GALLIUM-dotatate PET scan in a patient with a history of a phaeochromocytoma: Unsuspected uptake in papillary thyroid carcinoma metastases. J Neuroendocrinol 2021; 33:e12964. [PMID: 33754388 DOI: 10.1111/jne.12964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/21/2021] [Accepted: 03/05/2021] [Indexed: 11/29/2022]
Abstract
Scanning for somatostatin receptors using 68 Ga-dotatate positron emission tomography with co-registration with computed tomography or magnetic resonance imaging is now in widespread use for the identification of neuroendocrine tumours, phaeochromocytomas, and paragangliomas and their metastases. We present a case where a patient with a phaeochromocytoma showed uptake in her neck considered diagnostic of a head-and-neck paraganglioma, which was subsequently confirmed to be a metastatic papillary thyroid carcinoma. We alert clinicians to such falsely-identified tumours using this extensively used imaging technique.
Collapse
Affiliation(s)
- Daniel Moffat
- Department of Neurosurgery, Barts and the London NHS Trust, London, UK
| | - Polly Richards
- Department of Radiology, Barts and the London NHS Trust, London, UK
| | - Tom R Kurzawinski
- Department of Endocrine Surgery, University College Hospital, London, UK
| | - Sameer Khan
- Department of Nuclear Medicine, Hammersmith Hospital, London, UK
| | - Bernard Khoo
- Department of Endocrinology, Royal Free Hospital, London, UK
- ENETs Centre of Excellence, Royal Free Hospital, London, UK
| | - Ashley Grossman
- ENETs Centre of Excellence, Royal Free Hospital, London, UK
- Green Templeton College, University of Oxford, London, UK
| |
Collapse
|
37
|
Anderson RC, Velez EM, Desai B, Jadvar H. Management Impact of 68Ga-DOTATATE PET/CT in Neuroendocrine Tumors. Nucl Med Mol Imaging 2021; 55:31-37. [PMID: 33643487 DOI: 10.1007/s13139-020-00677-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/08/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose The goal of our retrospective single tertiary academic medical center investigation was to examine the added diagnostic value and clinical impact of 68Ga-DOTATATE PET/CT in the therapeutic management of patients with neuroendocrine tumors (NETs). Methods Imaging database was queried for all "PET-DOTATATE" examinations performed at our tertiary care academic institution using MONTAGE™. The patient's clinical history and recent prior imaging were reviewed. The additional diagnostic value and clinical management impact of 68Ga-DOTATATE were assessed through retrospective chart review. Results A total of 81 68Ga-DOTATATE PET/CT scans in 74 patients were found, and 11 patients were excluded from analysis as they had no prior imaging available for comparison, with resultant analysis cohort of 63 patients. Six patients had 2 or more 68Ga-DOTATATE PET/CT examinations. The most common primary diagnosis was undifferentiated NET (63.5%), followed by carcinoid (27.0%), paraganglioma (4.8%), insulinoma (3.2%), and pheochromocytoma (1.6%). The primary sites of disease from the most to the least common were the pancreas (36.5%), small bowel (22.2%), unknown primary (15.9%), lung (6.3%), large bowel (6.3%), and mesentery (4.8%), and other locations accounted for 7.9%. In patients who had prior imaging available for comparison, there were new lesions identified on 68Ga-DOTATATE PET/CT in 21 patients (33.3%) that were not identified on other prior imaging modalities. Of these patients, 5 underwent subsequent MRI and 1 had a repeat 68Ga-DOTATATE PET/CT to further characterize new lesions seen. Moreover, 15 patients (23.8%) had a change in treatment plan, including altering medical therapy in 9 patients, change in planned extent of surgical management in 5 patients, and cancelation of a planned primary tumor resection in 1 patient with metastatic disease. Conclusion Our retrospective cohort demonstrated that 68Ga-DOTATATE PET/CT improves lesion detection over conventional imaging in 33.3% and impacts the therapeutic management in 23.8% of patients with NET.
Collapse
Affiliation(s)
- Redmond-Craig Anderson
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA 90033 USA
| | - Erik M Velez
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA 90033 USA
| | - Bhushan Desai
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA 90033 USA
| | - Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar St., CSC 102, Los Angeles, CA 90033 USA
| |
Collapse
|
38
|
Bertani E, Collamati F, Colandrea M, Faccini R, Fazio N, Ferrari ME, Fischetti M, Fumagalli Romario U, Funicelli L, De Simoni M, Mancini-Terracciano C, Mirabelli R, Morganti S, Papi S, Pisa E, Solfaroli-Camillocci E, Spada F, Cremonesi M, Grana CM. First Ex Vivo Results of β --Radioguided Surgery in Small Intestine Neuroendocrine Tumors with 90Y-DOTATOC. Cancer Biother Radiopharm 2021; 36:397-406. [PMID: 33601932 DOI: 10.1089/cbr.2020.4487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: In neuroendocrine tumor (NET), complete surgery could better the prognosis. Radioguided surgery (RGS) with β--radioisotopes is a novel approach focused on developing a new probe that, detecting electrons and operating with low background, provides a clearer delineation of the lesions with low radiation exposition for surgeons. As a first step to validate this procedure, ex vivo specimens of tumors expressing somatostatin receptors, as small intestine neuroendocrine tumor (SI-NET), were tested. Materials and Methods: SI-NET presents a high uptake of a beta-emitting radiotracer, 90Y-DOTATOC. Five SI-NET patients were enrolled after performing a 68Ga-DOTATOC positron emission tomography/computed tomography (CT) and a CT enterography; 24 h before surgery, they received 5 mCi of 90Y-DOTATOC. Results: Surgery was performed as routine. Tumors and surrounding tissue were sectioned in different samples and examined ex vivo with the beta-detecting probe. All the tumor samples showed high counts of radioactivity that was up to a factor of 18 times higher than the corresponding cutoff value, with a sensitivity of 96% and a specificity of 100%. Conclusions: These first ex vivo RGS tests showed that this probe can discriminate very effectively between tumor and healthy tissues by the administration of low activities of 90Y-DOTATOC, allowing more precise surgery.
Collapse
Affiliation(s)
- Emilio Bertani
- Division of Digestive Surgery, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | | | - Marzia Colandrea
- Division of Nuclear Medicine, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Riccardo Faccini
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Dipartimento di Fisica, Università di Roma Sapienza, Roma, Italy
| | - Nicola Fazio
- Division of Gastrointestinal and Neuroendocrine Tumors Medical Treatment, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Mahila E Ferrari
- Medical Physics, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Marta Fischetti
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Dipartimento di Scienze di Base Applicate per l'Ingegneria, Sapienza Università di Roma, Roma, Italy
| | | | - Luigi Funicelli
- Division of Radiology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Micol De Simoni
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Dipartimento di Fisica, Università di Roma Sapienza, Roma, Italy
| | - Carlo Mancini-Terracciano
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Dipartimento di Fisica, Università di Roma Sapienza, Roma, Italy
| | - Riccardo Mirabelli
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Dipartimento di Fisica, Università di Roma Sapienza, Roma, Italy
| | - Silvio Morganti
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy
| | - Stefano Papi
- Division of Nuclear Medicine, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Eleonora Pisa
- Division of Pathology, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Elena Solfaroli-Camillocci
- Sezione di Roma, Istituto Nazionale di Fisica Nucleare, Roma, Italy.,Scuola di specializzazione in Fisica Medica, Sapienza Università di Roma, Roma, Italy
| | - Francesca Spada
- Division of Gastrointestinal and Neuroendocrine Tumors Medical Treatment, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Marta Cremonesi
- Medical Physics, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| | - Chiara M Grana
- Division of Nuclear Medicine, Istituto Europeo di Oncologia, IRCCS, Milano, Italy
| |
Collapse
|
39
|
Aloj L, Attili B, Lau D, Caraco C, Lechermann LM, Mendichovszky IA, Harper I, Cheow H, Casey RT, Sala E, Gilbert FJ, Gallagher FA. The emerging role of cell surface receptor and protein binding radiopharmaceuticals in cancer diagnostics and therapy. Nucl Med Biol 2021; 92:53-64. [PMID: 32563612 DOI: 10.1016/j.nucmedbio.2020.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022]
Abstract
Targeting specific cell membrane markers for both diagnostic imaging and radionuclide therapy is a rapidly evolving field in cancer research. Some of these applications have now found a role in routine clinical practice and have been shown to have a significant impact on patient management. Several molecular targets are being investigated in ongoing clinical trials and show promise for future implementation. Advancements in molecular biology have facilitated the identification of new cancer-specific targets for radiopharmaceutical development.
Collapse
Affiliation(s)
- Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom.
| | - Bala Attili
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Corradina Caraco
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Laura M Lechermann
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Iosif A Mendichovszky
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Ines Harper
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Heok Cheow
- Department of Nuclear Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ruth T Casey
- Department of Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Evis Sala
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Fiona J Gilbert
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| |
Collapse
|
40
|
Zhang YQ, Wen JX, Luo RK, Yuan HX, Wang WP. Imaging and Contrast-enhanced Ultrasound features of duodenal neuroendocrine tumor: A case report. Clin Hemorheol Microcirc 2020; 76:27-32. [PMID: 32538825 DOI: 10.3233/ch-200816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Duodenal neuroendocrine tumors are rare neoplasms arising from endocrine cells. Here we present a case of 32-year-old woman with Duodenal neuroendocrine tumors, report the imaging and contrast-enhanced Ultrasound (CEUS) features and review previous literatures of neuroendocrine tumors, which may be valuable for the differential diagnosis of duodenal neoplasms.
Collapse
Affiliation(s)
- Yan-Qun Zhang
- Department of Ultrasound, Xiamen Branch, Zhongshan Hospital of Fudan University, Xiamen, Fujian Province, China.,Department of Ultrasound, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jie-Xian Wen
- Department of Ultrasound, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Rong-Kui Luo
- Department of Pathology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Hai-Xia Yuan
- Department of Ultrasound, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Wen-Ping Wang
- Department of Ultrasound, Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
41
|
Jo H, Park Y, Kim J, Kwon H, Kim T, Lee J, Pyun JC, Lee M, Yun M. Elevated miR-16-5p induces somatostatin receptor 2 expression in neuroendocrine tumor cells. PLoS One 2020; 15:e0240107. [PMID: 33045023 PMCID: PMC7549806 DOI: 10.1371/journal.pone.0240107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/18/2020] [Indexed: 01/19/2023] Open
Abstract
Somatostatin analogs, which are used to treat neuroendocrine tumors, inhibit hormone secretion or promote tumor shrinkage; however, their efficacy varies between patients, possibly because of differential expression of somatostatin receptors (SSTRs) in tumors. In this study, we evaluated the regulatory mechanism underlying the expression of SSTR2, the main octreotide target. Thirty miRNAs were found to be dysregulated in neuroendocrine cells (INS-1 cells) incubated with octreotide compared to that in placebo-treated cells. Among the upregulated miRNAs, miR-16-5p was elevated after short-term octreotide treatment. We conducted in vitro experiments to determine whether the expression of miR-16-5p was associated with the regulation of SSTR2 expression and affected octreotide sensitivity in INS-1 cells. Overexpression of miR-16-5p by transfected mimics induced upregulation of SSTR2 expression. Additionally, the expression of miR-16-5p further enhanced octreotide-induced reduction in cell proliferation in both two- and three-dimensional culture of INS-1 cells. Thus, our results reveal the mechanism underlying SSTR2 expression regulation and may aid in developing therapeutic approaches for enhancing the response to octreotide, particularly in patients unresponsive to SSTR2-targeted somatostatin analog treatment.
Collapse
Affiliation(s)
- HanHee Jo
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yusun Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Jisu Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Hyeonjeong Kwon
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Taehun Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
| | - JongSook Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, Seoul, South Korea
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, South Korea
- * E-mail: (ML); (MY)
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- * E-mail: (ML); (MY)
| |
Collapse
|
42
|
Hu K, Xie L, Hanyu M, Zhang Y, Li L, Ma X, Nagatsu K, Suzuki H, Wang W, Zhang MR. Harnessing the PD-L1 interface peptide for positron emission tomography imaging of the PD-1 immune checkpoint. RSC Chem Biol 2020; 1:214-224. [PMID: 34458761 PMCID: PMC8341843 DOI: 10.1039/d0cb00070a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Interface peptides that mediate protein-protein interactions (PPI) are a class of important lead compounds for designing PPI inhibitors. However, their potential as precursors for radiotracers has never been exploited. Here we report that the interface peptides from programmed death-ligand 1 (PD-L1) can be used in positron emission tomography (PET) imaging of programmed cell death 1 (PD-1) with high accuracy and sensitivity. Moreover, the performance differentiation between murine PD-L1 derived interface peptide (mPep-1) and human PD-L1 derived interface peptide (hPep-1) as PET tracers for PD-1 unveiled an unprecedented role of a non-critical residue in target binding, highlighting the significance of PET imaging as a companion diagnostic in drug development. Collectively, this study not only provided a first-of-its-kind peptide-based PET tracer for PD-1 but also conveyed a unique paradigm for developing imaging agents for highly challenging protein targets, which could be used to identify other protein biomarkers involved in the PPI networks.
Collapse
Affiliation(s)
- Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Masayuki Hanyu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Lingyun Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 100081 P. R. China
| | - Xiaohui Ma
- Department of Vascular Surgery, General Hospital of People's Liberation Army Beijing 100853 P. R. China
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Hisashi Suzuki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| | - Weizhi Wang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 100081 P. R. China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology Chiba, 263-8555 Japan
| |
Collapse
|
43
|
Abstract
CLINICAL/METHODICAL ISSUE Conventional imaging tests like computed tomography (CT) cannot visualize somatostatin receptor (SSTR) expression on the tumor cell surface. STANDARD RADIOLOGICAL METHODS For imaging of SSTR-expressing tumors conventional morphological imaging tests such as CT or magnetic resonance imaging (MRI) are employed. METHODICAL INNOVATIONS Molecular imaging of SSTR expression on the tumor cell surface, in particular by using (whole body) single photon emission computed tomography (SPECT) and positron emission tomography (PET), are considered the current standard of care. Only the use of CT enables for exact localization of putative sites of disease (hybrid imaging). PERFORMANCE Hybrid SPECT/CT and PET/CT are of utmost importance for staging and monitoring of treatment efficacy. SSTR-PET is superior to SPECT and the PET radiotracer 68Ga-DOTATATE has been approved in multiple countries. In addition, SSTR positivity revealed by SPECT or PET pave the way for a peptide receptor radionuclide therapy (PRRT). Such a theranostic approach enables for systemic or locoregional radiation with β‑emitting radionuclides, which are linked to the identical amino acid peptide used for PET or SPECT imaging. The prospective, randomized Netter‑1 trial has shown significant benefit for patients receiving PRRT. ACHIEVEMENTS A combined use of conventional and functional imaging tests is superior to conventional imaging alone and allows for identification of suitable candidates for a theranostic approach. PRACTICAL RECOMMENDATIONS In case of clinical suspicion or after having obtained histological evidence, hybrid SSTR-SPECT/CT or -PET/CT should be performed, preferably in a dedicated molecular imaging center.
Collapse
|
44
|
Li D, Minnix M, Allen R, Bading J, Chea J, Wong P, Bowles N, Poku E, Shively JE. Preclinical PET Imaging of NTSR-1-Positive Tumors with 64Cu- and 68Ga-DOTA-Neurotensin Analogs and Therapy with an 225Ac-DOTA-Neurotensin Analog. Cancer Biother Radiopharm 2020; 36:651-661. [PMID: 32822229 DOI: 10.1089/cbr.2020.3926] [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: 10/23/2022] Open
Abstract
Background: The aim of the study was to perform PET imaging and radiotherapy with a novel neurotensin derivative for neurotensin receptor 1 (NTSR-1)-positive tumors in an animal model. Materials and Methods: A di-DOTA analog of NT(6-13) with three unnatural amino acids was synthesized and radiolabeled with either 64Cu or 68Ga and tested for serum stability and tumor imaging in mice bearing NTSR-1-positive PC3, and HT29 xenografts. A dose-response therapy study was performed with 18.5, 37, and 74 kBq of 225Ac-di-DOTA-α,ɛ-Lys-NT(6-13). Results: 68Ga-di-DOTA-α,ɛ-Lys-NT(6-13) was >99% stable in serum for 48 h, had an IC50 of 5 nM using 125I labeled NT(8-13) for binding to HT-29 cells, and high uptake in tumor models expressing NTSR-1. 68Ga-di-DOTA-α,ɛ-Lys-NT(6-13) had an average %ID/g (n = 4) at 2 h of 4.0 for tumor, 0.5 for blood, 12.0 for kidney, and <1 for other tissues, resulting in a favorable T/B of 8. Mean survivals of tumor-bearing mice treated with 18.5 or 37 kBq of 225Ac-di-DOTA-α,ɛ-Lys-NT(6-13) were 81 and 93 d, respectively, versus 53 d for controls. Whole-body toxicity was seen for the 74 kBq dose. Conclusions: Based on the results of the animal model, di-DOTA-α,ɛ-Lys-NT(6-13) is a useful imaging agent for NTSR-1-positive tumors when radiolabeled with 68Ga, and when radiolabeled with 225Ac, a potent therapeutic agent.
Collapse
Affiliation(s)
- Daneng Li
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Megan Minnix
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Rebecca Allen
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - James Bading
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Junie Chea
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - Patty Wong
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Nicole Bowles
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - Erasmus Poku
- Radiopharmacy, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - John E Shively
- Deparment of Molecular Imaging and Therapy, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
45
|
Czernin J, Sonni I, Razmaria A, Calais J. The Future of Nuclear Medicine as an Independent Specialty. J Nucl Med 2020; 60:3S-12S. [PMID: 31481589 DOI: 10.2967/jnumed.118.220558] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 07/06/2019] [Indexed: 02/07/2023] Open
Abstract
In this article, we provide an overview of established and emerging conventional nuclear medicine and PET imaging biomarkers, as the diagnostic nuclear medicine portfolio is rapidly expanding. Next, we review briefly nuclear theranostic approaches that have already entered or are about to enter clinical routine. Using some approximations and taking into account emerging applications, we also provide some simplified business forecasts for nuclear theranostics. We argue that an optimistic outlook by the nuclear medicine community is crucial to the growth of the specialty and emphasize the urgent need for training adaptations.
Collapse
Affiliation(s)
- Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Ida Sonni
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Aria Razmaria
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
46
|
Beyer T, Bidaut L, Dickson J, Kachelriess M, Kiessling F, Leitgeb R, Ma J, Shiyam Sundar LK, Theek B, Mawlawi O. What scans we will read: imaging instrumentation trends in clinical oncology. Cancer Imaging 2020; 20:38. [PMID: 32517801 PMCID: PMC7285725 DOI: 10.1186/s40644-020-00312-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Oncological diseases account for a significant portion of the burden on public healthcare systems with associated costs driven primarily by complex and long-lasting therapies. Through the visualization of patient-specific morphology and functional-molecular pathways, cancerous tissue can be detected and characterized non-invasively, so as to provide referring oncologists with essential information to support therapy management decisions. Following the onset of stand-alone anatomical and functional imaging, we witness a push towards integrating molecular image information through various methods, including anato-metabolic imaging (e.g., PET/CT), advanced MRI, optical or ultrasound imaging.This perspective paper highlights a number of key technological and methodological advances in imaging instrumentation related to anatomical, functional, molecular medicine and hybrid imaging, that is understood as the hardware-based combination of complementary anatomical and molecular imaging. These include novel detector technologies for ionizing radiation used in CT and nuclear medicine imaging, and novel system developments in MRI and optical as well as opto-acoustic imaging. We will also highlight new data processing methods for improved non-invasive tissue characterization. Following a general introduction to the role of imaging in oncology patient management we introduce imaging methods with well-defined clinical applications and potential for clinical translation. For each modality, we report first on the status quo and, then point to perceived technological and methodological advances in a subsequent status go section. Considering the breadth and dynamics of these developments, this perspective ends with a critical reflection on where the authors, with the majority of them being imaging experts with a background in physics and engineering, believe imaging methods will be in a few years from now.Overall, methodological and technological medical imaging advances are geared towards increased image contrast, the derivation of reproducible quantitative parameters, an increase in volume sensitivity and a reduction in overall examination time. To ensure full translation to the clinic, this progress in technologies and instrumentation is complemented by advances in relevant acquisition and image-processing protocols and improved data analysis. To this end, we should accept diagnostic images as "data", and - through the wider adoption of advanced analysis, including machine learning approaches and a "big data" concept - move to the next stage of non-invasive tumour phenotyping. The scans we will be reading in 10 years from now will likely be composed of highly diverse multi-dimensional data from multiple sources, which mandate the use of advanced and interactive visualization and analysis platforms powered by Artificial Intelligence (AI) for real-time data handling by cross-specialty clinical experts with a domain knowledge that will need to go beyond that of plain imaging.
Collapse
Affiliation(s)
- Thomas Beyer
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Währinger Gürtel 18-20/4L, 1090, Vienna, Austria.
| | - Luc Bidaut
- College of Science, University of Lincoln, Lincoln, UK
| | - John Dickson
- Institute of Nuclear Medicine, University College London Hospital, London, UK
| | - Marc Kachelriess
- Division of X-ray imaging and CT, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, DE, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074, Aachen, DE, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Am Fallturm 1, 28359, Bremen, DE, Germany
| | - Rainer Leitgeb
- Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, AT, Austria
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lalith Kumar Shiyam Sundar
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Währinger Gürtel 18-20/4L, 1090, Vienna, Austria
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074, Aachen, DE, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Am Fallturm 1, 28359, Bremen, DE, Germany
| | - Osama Mawlawi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
47
|
Kollenda SA, Klose J, Knuschke T, Sokolova V, Schmitz J, Staniszewska M, Costa PF, Herrmann K, Westendorf AM, Fendler WP, Epple M. In vivo biodistribution of calcium phosphate nanoparticles after intravascular, intramuscular, intratumoral, and soft tissue administration in mice investigated by small animal PET/CT. Acta Biomater 2020; 109:244-253. [PMID: 32251787 DOI: 10.1016/j.actbio.2020.03.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
Calcium phosphate nanoparticles were covalently surface-functionalized with the ligand DOTA and loaded with the radioisotope 68Ga. The biodistribution of such 68Ga-labelled nanoparticles was followed in vivo in mice by positron emission tomography in combination with computer tomography (PET-CT). The biodistribution of 68Ga-labelled nanoparticles was compared for different application routes: intravenous, intramuscular, intratumoral, and into soft tissue. The particle distribution was measured in vivo by PET-CT after 5 min, 15 min, 30 min, 1 h, 2 h, and 4 h, and ex vivo after 5 h. After intravenous injection (tail vein), the nanoparticles rapidly entered the lungs with later redistribution into liver and spleen. The nanoparticles remained mostly at the injection site following intramuscular, intratumoral, or soft tissue application, with less than 10 percent being mobilized into the blood stream. STATEMENT OF SIGNIFICANCE: The in vivo biodistribution of DOTA-terminated calcium phosphate nanoparticles was followed by PET/CT. To our knowledge, this is the first study of this kind. Four different application routes of clinical relevance were pursued: Intravascular, intramuscular, intratumoral, and into soft tissue. Given the high importance of calcium phosphate as biomaterial and for nanoparticular drug delivery and immunization, this is most important to assess the biofate of calcium phosphate nanoparticles for therapeutic application and also judge biodistribution of nanoscopic calcium phosphate ceramics, including debris from endoprostheses and related implants.
Collapse
Affiliation(s)
- Sebastian A Kollenda
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Jasmin Klose
- Department of Nuclear Medicine, University Hospital and German Cancer Consortium (DKTK) Partner Site Essen, University of Duisburg-Essen, Essen, Germany
| | - Torben Knuschke
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Viktoriya Sokolova
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany
| | - Jochen Schmitz
- Department of Radiopharmacy and Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Magdalena Staniszewska
- Department of Nuclear Medicine, University Hospital and German Cancer Consortium (DKTK) Partner Site Essen, University of Duisburg-Essen, Essen, Germany
| | - Pedro Fragoso Costa
- Department of Nuclear Medicine, University Hospital and German Cancer Consortium (DKTK) Partner Site Essen, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital and German Cancer Consortium (DKTK) Partner Site Essen, University of Duisburg-Essen, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital and German Cancer Consortium (DKTK) Partner Site Essen, University of Duisburg-Essen, Essen, Germany.
| | - Matthias Epple
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
48
|
Hibi T, Rela M, Eason JD, Line PD, Fung J, Sakamoto S, Selzner N, Man K, Ghobrial RM, Sapisochin G. Liver Transplantation for Colorectal and Neuroendocrine Liver Metastases and Hepatoblastoma. Working Group Report From the ILTS Transplant Oncology Consensus Conference. Transplantation 2020; 104:1131-1135. [PMID: 32217939 DOI: 10.1097/tp.0000000000003118] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver transplantation (LT) for unresectable colorectal liver metastases has long been abandoned because of dismal prognoses. After the dark ages, advances in chemotherapy and diagnostic imaging have enabled strict patient selection, and the pioneering study from the Oslo group has contributed to the substantial progress in this field. For unresectable neuroendocrine liver metastases, LT for patients who met the Milan criteria was able to achieve excellent long-term outcomes. The guidelines further adopted in the United States and Europe were based on these criteria. For hepatoblastoma, patients with unresectable and borderline-resectable disease are considered good candidates for LT; however, the indications are yet to be defined. In the budding era of transplant oncology, it is critically important to recognize the current status and unsolved questions for each disease entity. These guidelines were developed to serve as a beacon of light for optimal patient selection for LT and set the stage for future basic and clinical studies.
Collapse
Affiliation(s)
- Taizo Hibi
- Department of Pediatric Surgery and Transplantation, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Mohamed Rela
- Dr. Rela Institute & Medical Centre, Chennai, India
| | - James D Eason
- James D. Eason Transplant Institute, Methodist/University of Tennessee Hospital, Memphis, TN
| | - Pål-Dag Line
- Department of Transplantation Medicine, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Fung
- University of Chicago Medicine Transplant Institute, Chicago, IL
| | - Seisuke Sakamoto
- Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Nazia Selzner
- Multi-Organ Transplant, Department of Medicine, University of Toronto, Toronto, Canada
| | - Kwan Man
- Department of Surgery, University of Hong Kong, Hong Kong, China
| | - R Mark Ghobrial
- Sherrie and Alan Conover Center for Liver Disease & Transplantation, and Department of Surgery, Weill Cornell Medical College, Houston, TX
| | - Gonzalo Sapisochin
- Multi-Organ Transplant, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Ectopic adrenocorticotropic hormone (ACTH)-secreting tumors are commonly small, yet they often lead to fulminant forms of Cushing syndrome. High-resolution functional imaging modalities, such as [Ga]-DOTATATE, have been recently introduced in clinical practice for the identification of neuroendocrine tumors. In this review, we focus on the performance of [Ga]-DOTATATE as a tool for localizing primary and metastatic sources of ectopic Cushing syndrome (ECS). RECENT FINDINGS Prompt surgical removal of ectopic ACTH-secreting tumors is the mainstay of therapy in patients with ECS. Detecting such tumors with conventional cross-sectional imaging is often unsuccessful, owing to their small size. [Ga]-DOTATATE has been approved in 2016 by the Federal Drug Administration for imaging well differentiated neuroendocrine tumors. Data regarding the performance of [Ga]-DOTATATE for detecting ectopic ACTH-secreting tumors remain limited, in part owing to the recent introduction of this imaging modality in clinical practice, and in part because of the low prevalence of ECS. Nevertheless, [Ga]-DOTATATE has been reported to be useful in identifying primary and metastatic ectopic ACTH-secreting lesions that were not apparent on other imaging studies, impacting the clinical care of many patients with ECS. SUMMARY [Ga]-DOTATATE-based imaging, which targets the somatostatin receptors abundantly expressed in neuroendocrine tumors, has generally high, although variable resolution in detecting the source(s) of ECS.
Collapse
Affiliation(s)
- Seda Grigoryan
- Department of Internal Medicine, Michigan State University, East Lasing
| | | | - Adina F Turcu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
50
|
Werner RA, Hänscheid H, Leal JP, Javadi MS, Higuchi T, Lodge MA, Buck AK, Pomper MG, Lapa C, Rowe SP. Impact of Tumor Burden on Quantitative [ 68Ga] DOTATOC Biodistribution. Mol Imaging Biol 2020; 21:790-798. [PMID: 30406512 DOI: 10.1007/s11307-018-1293-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE As has been previously reported, the somatostatin receptor (SSTR) imaging agent [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotate ([68Ga]DOTATATE) demonstrates lower uptake in normal organs in patients with a high neuroendocrine tumor (NET) burden. Given the higher SSTR affinity of [68Ga] DOTATATE, we aimed to quantitatively investigate the biodistribution of [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotide ([68Ga]DOTATOC) to determine a potential correlation between uptake in normal organs and NET burden. PROCEDURES Of the 44 included patients, 36/44 (82 %) patients demonstrated suspicious radiotracer uptake on [68Ga] DOTATOC positron emission tomography (PET)/X-ray computed tomography (CT). Volumes of interest (VOIs) were defined for tumor lesions and normal organs (spleen, liver, kidneys, adrenals). Mean body weight corrected standardized uptake value (SUVmean) for normal organs was assessed and was used to calculate the corresponding mean specific activity uptake (Upt: fraction of injected activity per kg of tissue). For the entire tumor burden, SUVmean, maximum standardized uptake value (SUVmax), and the total mass (TBM) was calculated and the decay corrected tumor fractional uptake (TBU) was assessed. A Spearman's rank correlation coefficient was used to determine the correlations between normal organ uptake and tumor burden. RESULTS The median SUVmean was 18.7 for the spleen (kidneys, 9.2; adrenals, 6.8; liver, 5.6). For tumor burden, the median values were SUVmean 6.9, SUVmax 35.5, TBM 42.6 g, and TBU 1.2 %. With increasing volume of distribution, represented by lean body mass and body surface area (BSA), Upt decreased in kidneys, liver, and adrenal glands and SUVmean increased in the spleen. Correlation improved only for both kidneys and adrenals when the influence of the tumor uptake on the activity available for organ uptake was taken into account by the factor 1/(1-TBU). TBU was neither predictive for SUVmean nor for Upt in any of the organs. The distribution of organ Upt vs. BSA/(1-TBU) were not different for patients with minor TBU (<3 %) vs. higher TBU (>7 %), indicating that the correlations observed in the present study are explainable by the body size effect. High tumor mass and uptake mitigated against G1 NET. CONCLUSIONS There is no significant impact on normal organ biodistribution with increasing tumor burden on [68Ga] DOTATOC PET/CT. Potential implications include increased normal organ dose with [177Lu-DOTA]0-D-Phe1-Tyr3-Octreotide and decreased absolute lesion detection with [68Ga] DOTATOC in high NET burden.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany.,The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Heribert Hänscheid
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jeffrey P Leal
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Mehrbod S Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin A Lodge
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA. .,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|