201
|
Maucherat B, Varmenot N, Fleury V, Senellart H, Rousseau C. Effective Management of 177Lu-DOTA0-Tyr3-Octreotate Extravasation. Clin Nucl Med 2021; 46:144-145. [PMID: 33323730 DOI: 10.1097/rlu.0000000000003452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Radiopharmaceutical extravasation is a known nuclear medicine adverse effect, mostly with no complication in case of diagnostic radiopharmaceutical. However, a therapeutic radiopharmaceutical extravasation may have clinical consequences and must be treated quickly and effectively. We report here a case of 177Lu-DOTA0-Tyr3-octreotate extravasation.
Collapse
|
202
|
Liberini V, Rampado O, Gallio E, De Santi B, Ceci F, Dionisi B, Thuillier P, Ciuffreda L, Piovesan A, Fioroni F, Versari A, Molinari F, Deandreis D. 68Ga-DOTATOC PET/CT-Based Radiomic Analysis and PRRT Outcome: A Preliminary Evaluation Based on an Exploratory Radiomic Analysis on Two Patients. Front Med (Lausanne) 2021; 7:601853. [PMID: 33575262 PMCID: PMC7870479 DOI: 10.3389/fmed.2020.601853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Aim: This work aims to evaluate whether the radiomic features extracted by 68Ga-DOTATOC-PET/CT of two patients are associated with the response to peptide receptor radionuclide therapy (PRRT) in patients affected by neuroendocrine tumor (NET). Methods: This is a pilot report in two NET patients who experienced a discordant response to PRRT (responder vs. non-responder) according to RECIST1.1. The patients presented with liver metastasis from the rectum and pancreas G3-NET, respectively. Whole-body total-lesion somatostatin receptor-expression (TLSREwb-50) and somatostatin receptor-expressing tumor volume (SRETV wb-50) were obtained in pre- and post-PRRT PET/CT. Radiomic analysis was performed, extracting 38 radiomic features (RFs) from the patients' lesions. The Mann–Whitney test was used to compare RFs in the responder patient vs. the non-responder patient. Pearson correlation and principal component analysis (PCA) were used to evaluate the correlation and independence of the different RFs. Results: TLSREwb-50 and SRETVwb-50 modifications correlate with RECIST1.1 response. A total of 28 RFs extracted on pre-therapy PET/CT showed significant differences between the two patients in the Mann–Whitney test (p < 0.05). A total of seven second-order features, with poor correlation with SUVmax and PET volume, were identified by the Pearson correlation matrix. Finally, the first two PCA principal components explain 83.8% of total variance. Conclusion: TLSREwb-50 and SRETVwb-50 are parameters that might be used to predict and to assess the PET response to PRRT. RFs might have a role in defining inter-patient heterogeneity and in the prediction of therapy response. It is important to implement future studies with larger and more homogeneous patient populations to confirm the efficacy of these biomarkers.
Collapse
Affiliation(s)
- Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Osvaldo Rampado
- Medical Physics Unit, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Elena Gallio
- Medical Physics Unit, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Bruno De Santi
- Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy
| | - Francesco Ceci
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Beatrice Dionisi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Philippe Thuillier
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy.,Department of Endocrinology, University Hospital of Brest, Brest, France
| | - Libero Ciuffreda
- Medical Oncology Division 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Alessandro Piovesan
- Division of Oncological Endocrinology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Federica Fioroni
- Medical Physics Unit, Azienda Unit Sanitaria Locale di Reggio Emilia - Istituto di Ricovero e Cura a Carattere Scientifico of Reggio Emilia, Reggio Emilia, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda Unit Sanitaria Locale di Reggio Emilia - Istituto di Ricovero e Cura a Carattere Scientifico of Reggio Emilia, Reggio Emilia, Italy
| | - Filippo Molinari
- Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Turin, Italy
| | - Désirée Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
203
|
HIF2alpha-Associated Pseudohypoxia Promotes Radioresistance in Pheochromocytoma: Insights from 3D Models. Cancers (Basel) 2021; 13:cancers13030385. [PMID: 33494435 PMCID: PMC7865577 DOI: 10.3390/cancers13030385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/30/2022] Open
Abstract
Pheochromocytomas and paragangliomas (PCCs/PGLs) are rare neuroendocrine tumors arising from chromaffin tissue located in the adrenal or ganglia of the sympathetic or parasympathetic nervous system. The treatment of non-resectable or metastatic PCCs/PGLs is still limited to palliative measures, including somatostatin type 2 receptor radionuclide therapy with [177Lu]Lu-DOTA-TATE as one of the most effective approaches to date. Nevertheless, the metabolic and molecular determinants of radiation response in PCCs/PGLs have not yet been characterized. This study investigates the effects of hypoxia-inducible factor 2 alpha (HIF2α) on the susceptibility of PCCs/PGLs to radiation treatments using spheroids grown from genetically engineered mouse pheochromocytoma (MPC) cells. The expression of Hif2α was associated with the significantly increased resistance of MPC spheroids to external X-ray irradiation and exposure to beta particle-emitting [177Lu]LuCl3 compared to Hif2α-deficient controls. Exposure to [177Lu]LuCl3 provided an increased long-term control of MPC spheroids compared to single-dose external X-ray irradiation. This study provides the first experimental evidence that HIF2α-associated pseudohypoxia contributes to a radioresistant phenotype of PCCs/PGLs. Furthermore, the external irradiation and [177Lu]LuCl3 exposure of MPC spheroids provide surrogate models for radiation treatments to further investigate the metabolic and molecular determinants of radiation responses in PCCs/PGLs and evaluate the effects of neo-adjuvant-in particular, radiosensitizing-treatments in combination with targeted radionuclide therapies.
Collapse
|
204
|
D'Amico G, Uso TD, Del Prete L, Hashimoto K, Aucejo FN, Fujiki M, Eghtesad B, Sasaki K, David Kwon CH, Miller CM, Quintini C. Neuroendocrine liver metastases: The role of liver transplantation. Transplant Rev (Orlando) 2021; 35:100595. [PMID: 33548685 DOI: 10.1016/j.trre.2021.100595] [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: 10/23/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW Neuroendocrine tumor (NET) metastasis localized to the liver is an accepted indication for liver transplantation as such tumors have a low biological aggressiveness in terms of malignancy and are slow growing. RECENT FINDINGS The long-term results are comparable with and in some cases even better than those of transplantations performed for primary liver cancer. However, compared with nonmalignant conditions, neuroendocrine liver metastasis (NELM) may result in an inferior outcome of transplantation. In the face of the scarcity of donated organs and recent improved results of non-surgical treatment for NELM, controversy over patient selection and timing for liver transplantation continues. SUMMARY In this review, we provide an overview of the diagnostic work-up and selection criteria of patients with NELM being considered for liver transplantation. Thereafter, we provide a critical analysis of the reported outcomes of OLT.
Collapse
Affiliation(s)
- Giuseppe D'Amico
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Teresa Diago Uso
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Luca Del Prete
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Koji Hashimoto
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Federico N Aucejo
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Masato Fujiki
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bijan Eghtesad
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kazunari Sasaki
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Choon H David Kwon
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Charles M Miller
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cristiano Quintini
- Transplantation Center, Department of Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
205
|
Hänscheid H, Hartrampf PE, Schirbel A, Buck AK, Lapa C. Intraindividual comparison of [ 177Lu]Lu-DOTA-EB-TATE and [ 177Lu]Lu-DOTA-TOC. Eur J Nucl Med Mol Imaging 2021; 48:2566-2572. [PMID: 33452632 PMCID: PMC8241641 DOI: 10.1007/s00259-020-05177-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/20/2020] [Indexed: 11/04/2022]
Abstract
Purpose The radiolabelled somatostatin analogue [177Lu]Lu-DOTA-EB-TATE binds to albumin via Evans blue, thereby increasing the residence time in the blood and potentially allowing more therapeutic agent to be absorbed into the target tissue during peptide receptor radionuclide therapy. It was tested in selected patients whether the substance is superior to [177Lu]Lu-DOTA-TOC. Methods Activity kinetics in organs and tumours after [177Lu]Lu-DOTA-EB-TATE and [177Lu]Lu-DOTA-TOC were compared intraindividually in five patients with progressive somatostatin receptor-positive disease scheduled for radionuclide therapy. Results In comparison to [177Lu]Lu-DOTA-TOC, tumour doses per administered activity were higher for [177Lu]Lu-DOTA-EB-TATE in 4 of 5 patients (median ratio: 1.7; range: 0.9 to 3.9), kidney doses (median ratio: 3.2; range: 1.6 to 9.8) as well as spleen doses (median ratio: 4.7; range 1.2 to 6.2) in all patients, and liver doses in 3 of 4 evaluable patients (median ratio: 4.0; range: 0.7 to 4.9). The tumour to critical organs absorbed dose ratios were higher after [177Lu]Lu-DOTA-TOC in 4 of 5 patients. Conclusions Prior to a treatment with [177Lu]Lu-DOTA-EB-TATE, it should be assessed individually whether the compound is superior to established substances.
Collapse
Affiliation(s)
- Heribert Hänscheid
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.,Department of Nuclear Medicine, University Hospital Augsburg, Augsburg, Germany
| |
Collapse
|
206
|
Raymond LM, Korzun T, Kardosh A, Kolbeck KJ, Pommier R, Mittra ES. The State of Peptide Receptor Radionuclide Therapy and Its Sequencing among Current Therapeutic Options for Gastroenteropancreatic Neuroendocrine Tumors. Neuroendocrinology 2021; 111:1086-1098. [PMID: 33744879 DOI: 10.1159/000516015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 03/17/2021] [Indexed: 11/19/2022]
Abstract
Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are the most common form of neuroendocrine neoplasia, but there is no current consensus for the sequencing of approved therapies, particularly with respect to peptide receptor radionuclide therapy (PRRT). This comprehensive review evaluates the data supporting approved therapies for GEP-NETs and recommendations for therapeutic sequencing with a focus on how PRRT currently fits within sequencing algorithms. The current recommendations for PRRT sequencing restrict its use to metastatic, inoperable, progressive midgut NETs; however, this may change with emerging data to suggest that PRRT might be beneficial as neoadjuvant therapy for inoperable tumors, is more tolerable than other treatment modalities following first-line standard dose somatostatin analogs, and can be used as salvage therapy after disease relapse following prior successful cycles of PRRT. PRRT has also been shown to reduce tumor burden, improve quality of life, and prolong the time to disease progression in a broad spectrum of patients with GEP-NETs. As the various potential benefits of PRRT in GEP-NET therapy continues to expand, it is necessary to review and critically evaluate our treatment algorithms for GEP-NETs.
Collapse
Affiliation(s)
- Lauren M Raymond
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA,
| | - Tetiana Korzun
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Adel Kardosh
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hematology/Medical Oncology, Department of Internal Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Kenneth J Kolbeck
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Interventional Radiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rodney Pommier
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Division of Surgical Oncology, Department of Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Erik S Mittra
- School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Division of Nuclear Medicine & Molecular Imaging, Department of Diagnostic Radiology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
207
|
Patel TK, Adhikari N, Amin SA, Biswas S, Jha T, Ghosh B. Small molecule drug conjugates (SMDCs): an emerging strategy for anticancer drug design and discovery. NEW J CHEM 2021. [DOI: 10.1039/d0nj04134c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms of how SMDCs work. Small molecule drugs are conjugated with the targeted ligand using pH sensitive linkers which allow the drug molecule to get released at lower lysosomal pH. It helps to accumulate the chemotherapeutic agents to be localized in the tumor environment upon cleaving of the pH-labile bonds.
Collapse
Affiliation(s)
- Tarun Kumar Patel
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Nilanjan Adhikari
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| |
Collapse
|
208
|
Haddad T, Fard-Esfahani A, Vali R. A review of pediatric neuroendocrine tumors, their detection, and treatment by radioisotopes. Nucl Med Commun 2021; 42:21-31. [PMID: 33044400 DOI: 10.1097/mnm.0000000000001305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroendocrine tumors (NETs) are rare in childhood. Neuroblastoma is the most common pediatric extracranial solid tumor, occurring >90% in children younger than 5 years of age. Pheochromocytoma and paraganglioma are rare NETs, causing hypertension in 0.5-2% of hypertensive children. Gastroenteropancreatic NETs can occur in children and are classified into carcinoids and pancreatic tumors. Nuclear medicine procedures have an essential role both in the diagnosis and treatment of NETs. Metaiodobenzylguanidine (MIBG) labeled with radioiodine has a well-established role in diagnosis as well as therapeutic management of the neuroblastoma group of diseases. During recent decades, establishing the abundant expression of somatostatin receptors by NETs first led to scintigraphy with somatostatin analogs (i.e. Tc/In-octreotide) and, later, with the emergence of positron-emitting labeled agents (i.e. Ga-DOTATATE/DOTATOC/DOTANOC) PET scans with significantly higher detection efficiency became available. Therapy with somatostatin analogs labeled with beta emitters such as Lu-177 and Y-90, known as peptide receptor radionuclide therapy, is a promising new option in the management of patients with inoperable or metastasized NETs. In this article, pediatric NETs are briefly reviewed and the role of radioactive agents in the detection and treatment of these tumors is discussed.
Collapse
Affiliation(s)
- Tara Haddad
- Diagnostic Imaging Department, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Armaghan Fard-Esfahani
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Vali
- Diagnostic Imaging Department, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
209
|
Abstract
Neuroendocrine tumors (NETs) are a heterogeneous group of tumors that originate in endocrine tissues throughout the body. Peptide receptor radionuclide therapy (PRRT) has emerged as a promising therapeutic option for patients with locally advanced and/or metastatic disease refractory to standard of care treatment. The landmark international phase III NETTER-1 trial led to the approval of 177Lu-DOTATATE (Lutathera) in the treatment of somatostatin receptor-positive gastroenteropancreatic NETs. Similarly, data from the multicenter, phase II Study IB12B led to the approval of meta-[131I]Iodo-Benzyl-Guanidine (I31I-MIBG) for treatment of iobenguane scan-positive, unresectable, locally advanced or metastatic pheochromocytoma or paraganglioma. With the clinical approval of these novel radiopharmaceuticals for managing select patients with NETs, additional studies are needed to refine patient selection, predict and assess therapy response, and optimize radiopharmaceutical delivery and clinical outcomes.
Collapse
Affiliation(s)
- Re-I Chin
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Francis S Wu
- Department of Radiology, St. Louis University, St. Louis, MO
| | - Yusuf Menda
- Department of Radiology, University of Iowa, Iowa City, IA
| | - Hyun Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
210
|
Könik A, O'Donoghue JA, Wahl RL, Graham MM, Van den Abbeele AD. Theranostics: The Role of Quantitative Nuclear Medicine Imaging. Semin Radiat Oncol 2021; 31:28-36. [PMID: 33246633 DOI: 10.1016/j.semradonc.2020.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Theranostics is a precision medicine discipline that integrates diagnostic nuclear medicine imaging with radionuclide therapy in a manner that provides both a tumor phenotype and personalized therapy to patients with cancer using radiopharmaceuticals aimed at the same target-specific biological pathway or receptor. The aim of quantitative nuclear medicine imaging is to plan the alpha or beta-emitting therapy based on an accurate 3-dimensional representation of the in-vivo distribution of radioactivity concentration within the tumor and normal organs/tissues in a noninvasive manner. In general, imaging may be either based on positron emission tomography (PET) or single photon emission computed tomography (SPECT) invariably in combination with X-ray CT (PET/CT; SPECT/CT) or, to a much lesser extent, MRI. PET and SPECT differ in terms of the radionuclides and physical processes that give rise to the emission of high energy photons, as well as the sets of technologies involved in their detection. Using a variety of standardized quantitative parameters, system calibration, patient preparation, imaging acquisition and reconstruction protocols, and image analysis protocols, an accurate quantification of the tracer distribution can be obtained, which helps prescribe the therapeutic dose for each patient.
Collapse
Affiliation(s)
- Arda Könik
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA.
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard L Wahl
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St. Louis, MO
| | - Michael M Graham
- Past Director of Nuclear Medicine, Roy J and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Annick D Van den Abbeele
- Department of Imaging, Dana-Farber Cancer Institute, Boston, MA; Division of Cancer Imaging, Mass General Brigham, Boston, MA; Dana-Farber Cancer Institute and Mass General Brigham, Boston, MA; Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA; Tumor Imaging Metrics Core, Dana-Farber/Harvard Cancer Center, Boston, MA
| |
Collapse
|
211
|
Vida Navas EM, Martínez Lorca A, Sancho Gutiérrez A, Sanz Gómez L, Navarro Martínez T, Grande Pulido E, Carrato Mena A, Gajate Borau P. Case Report: Re-Treatment With Lu-DOTATATE in Neuroendocrine Tumors. Front Endocrinol (Lausanne) 2021; 12:676973. [PMID: 33935979 PMCID: PMC8082310 DOI: 10.3389/fendo.2021.676973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/26/2021] [Indexed: 01/01/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) is an established treatment in advanced neuroendocrine tumors (NETs), which overexpressed somatostatin receptors. However, after progression there are a limited number of available treatments. We want to share a case report about a patient with a NET re-treated with 177Lu-DOTATATE and a literature review about salvage treatment with PRRT. We present a 26-year-old man who started with pelvic pain and after a biopsy of a retro-rectal mass observed in a magnetic resonance was diagnosed with an advanced neuroendocrine tumour. After progression to lanreotide, everolimus and sunitinib, treatment with 177Lu-DOTATATE was initiated, achieving an excellent response with a progression free survival (PFS) of 38 months. At the time of progression, re-treatment with 177Lu-DOTATATE was decided, showing a new partial response, which is currently stable after 15 months. The patient had not presented significant treatment-related toxicity. Although there are no randomized phase III trials or a consensus about the number or dose of cycles, there is evidence about the efficacy and low toxicity of salvage treatment with 177Lu-DOTATATE in NETs. Median progression-free survival ranges from 6 to 22 months. Toxicity is mostly hematologic (anemia and neutropenia), 4-7% grade 3/4.
Collapse
Affiliation(s)
- Elena María Vida Navas
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- *Correspondence: Elena María Vida Navas,
| | | | | | - Lucia Sanz Gómez
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | | | | | - Alfredo Carrato Mena
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Pablo Gajate Borau
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| |
Collapse
|
212
|
Konijnenberg M, Herrmann K, Kobe C, Verburg F, Hindorf C, Hustinx R, Lassmann M. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging 2021; 48:67-72. [PMID: 33057773 PMCID: PMC7835146 DOI: 10.1007/s00259-020-05038-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022]
Abstract
The EC Directive 2013/59/Euratom states in article 56 that exposures of target volumes in nuclear medicine treatments shall be individually planned and their delivery appropriately verified. The Directive also mentions that medical physics experts should always be appropriately involved in those treatments. Although it is obvious that, in nuclear medicine practice, every nuclear medicine physician and physicist should follow national rules and legislation, the EANM considered it necessary to provide guidance on how to interpret the Directive statements for nuclear medicine treatments.For this purpose, the EANM proposes to distinguish three levels in compliance to the optimization principle in the directive, inspired by the indication of levels in prescribing, recording and reporting of absorbed doses after radiotherapy defined by the International Commission on Radiation Units and Measurements (ICRU): Most nuclear medicine treatments currently applied in Europe are standardized. The minimum requirement for those treatments is ICRU level 1 ("activity-based prescription and patient-averaged dosimetry"), which is defined by administering the activity within 10% of the intended activity, typically according to the package insert or to the respective EANM guidelines, followed by verification of the therapy delivery, if applicable. Non-standardized treatments are essentially those in developmental phase or approved radiopharmaceuticals being used off-label with significantly (> 25% more than in the label) higher activities. These treatments should comply with ICRU level 2 ("activity-based prescription and patient-specific dosimetry"), which implies recording and reporting of the absorbed dose to organs at risk and optionally the absorbed dose to treatment regions. The EANM strongly encourages to foster research that eventually leads to treatment planning according to ICRU level 3 ("dosimetry-guided patient-specific prescription and verification"), whenever possible and relevant. Evidence for superiority of therapy prescription on basis of patient-specific dosimetry has not been obtained. However, the authors believe that a better understanding of therapy dosimetry, i.e. how much and where the energy is delivered, and radiobiology, i.e. radiation-related processes in tissues, are keys to the long-term improvement of our treatments.
Collapse
Affiliation(s)
- Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Frederik Verburg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Cecilia Hindorf
- Department of Nuclear Medicine Physics, Skåne University Hospital, Lund, Sweden
| | - Roland Hustinx
- Service de Médecine Nucléaire et d'Imagerie Oncologique, Département de Physique médicale, Université de Liège - GIGA-CRC in vivo imaging, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - Michael Lassmann
- Department of Nuclear Medicine, Universitätsklinikum Würzburg, Klinik und Poliklinik für Nuklearmedizin, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| |
Collapse
|
213
|
Hanaoka K, Miyaji N, Yoneyama H, Ogawa M, Maeda T, Sakaguchi K, Iimori T, Tsushima H. [Radiological Technology for Targeted Radionuclide Therapy]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2020; 76:1237-1247. [PMID: 33342942 DOI: 10.6009/jjrt.2020_jsrt_76.12.1237] [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: 11/11/2022]
Abstract
Targeted radioisotope therapy (TRT) is a radiotherapy using radioisotope or drug incorporating it and has been used as a treatment for selectively irradiating cancer cells. In recent years, interest in TRT has increased due to improvements in radionuclide production technology, development of new drugs and imaging modalities, and improvements in radiation technology. In order to enhance the effect of TRT, measurement of individual radiation doses to tumor tissue and organs at risk is important using highly quantitative nuclear medicine images. In this paper, we present a review of literature on optimization of TRT, which is a new research area from the perspective of radiation technology.
Collapse
Affiliation(s)
- Kohei Hanaoka
- Institute of Advanced Clinical Medicine, Kindai University
| | - Noriaki Miyaji
- Department of Nuclear Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research
| | - Hiroto Yoneyama
- Department of Radiological Technology, Kanazawa University Hospital
| | | | - Takamasa Maeda
- Radiological Technology Section, QST Hospital, National Institutes for Quantum and Radiological Science and Technology
| | | | | | - Hiroyuki Tsushima
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences
| |
Collapse
|
214
|
Efficacy of 177Lu-Dotatate Induction and Maintenance Therapy of Various Types of Neuroendocrine Tumors: A Phase II Registry Study. ACTA ACUST UNITED AC 2020; 28:115-127. [PMID: 33622997 PMCID: PMC7816182 DOI: 10.3390/curroncol28010015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) has been recently established as a treatment option for progressive gastro-entero-pancreatic neuroendocrine tumors (NETs) including four 200 mCi induction cycles. The purpose of this phase 2 trial is to expand use of PRRT to different types of NETs with the application of dose adjustment and evaluate value of maintenance therapy in patients who had disease control on induction therapy. Forty-seven PRRT naïve NET patients with different primary origin received 177Lu-DOTATATE induction therapy, ranging from 75 to 150 mCi per cycle, based on patients’ clinical status such as liver and renal function, extent of metastases, and previous therapies. Thirty-four patients underwent additional maintenance therapy (50–100 mCi per cycle) following induction course until they developed disease progression. The estimated median progression-free survival (PFS) was 36.1 months. The median PFS in our MNET subgroup was 47.7 months, which is markedly longer than NETTER-1 trial with median PFS of 28.4 months. The median PFS was significantly longer in patients who received PRRT as first-line treatment after disease progression on somatostatin analogs compared to patients who received other therapies first (p-value = 0.04). The total disease response rate (DRR) and disease control rate (DCR) was 32% and 85% based on RECIST 1.1 and 45% and 83% based on Choi criteria. This trial demonstrates longer PFS with the addition of low dose maintenance therapy to induction therapy compared to NETTER-1 trial that only included induction therapy. Also, we observed considerable efficacy of PRRT in various types of advanced NETs.
Collapse
|
215
|
Liberini V, Huellner MW, Grimaldi S, Finessi M, Thuillier P, Muni A, Pellerito RE, Papotti MG, Piovesan A, Arvat E, Deandreis D. The Challenge of Evaluating Response to Peptide Receptor Radionuclide Therapy in Gastroenteropancreatic Neuroendocrine Tumors: The Present and the Future. Diagnostics (Basel) 2020; 10:E1083. [PMID: 33322819 PMCID: PMC7763988 DOI: 10.3390/diagnostics10121083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The NETTER-1 study has proven peptide receptor radionuclide therapy (PRRT) to be one of the most effective therapeutic options for metastatic neuroendocrine tumors (NETs), improving progression-free survival and overall survival. However, PRRT response assessment is challenging and no consensus on methods and timing has yet been reached among experts in the field. This issue is owed to the suboptimal sensitivity and specificity of clinical biomarkers, limitations of morphological response criteria in slowly growing tumors and necrotic changes after therapy, a lack of standardized parameters and timing of functional imaging and the heterogeneity of PRRT protocols in the literature. The aim of this article is to review the most relevant current approaches for PRRT efficacy prediction and response assessment criteria in order to provide an overview of suitable tools for safe and efficacious PRRT.
Collapse
Affiliation(s)
- Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Serena Grimaldi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Monica Finessi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Philippe Thuillier
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Endocrinology, University Hospital of Brest, 29200 Brest, France
| | - Alfredo Muni
- Department of Nuclear Medicine, S.S. Biagio e Antonio e C. Arrigo Hospital, 15121 Alessandria, Italy;
| | | | - Mauro G. Papotti
- Pathology Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
- Department of Oncology, University of Turin at Molinette Hospital, 10126 Turin, Italy
| | - Alessandro Piovesan
- Department of Endocrinology, A. O. U. Città della Salute della Scienza of Turin, 10126 Turin, Italy;
| | - Emanuela Arvat
- Oncological Endocrinology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Désirée Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| |
Collapse
|
216
|
Calabrò D, Argalia G, Ambrosini V. Role of PET/CT and Therapy Management of Pancreatic Neuroendocrine Tumors. Diagnostics (Basel) 2020; 10:E1059. [PMID: 33297381 PMCID: PMC7762240 DOI: 10.3390/diagnostics10121059] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are heterogeneous neoplasms with neuroendocrine differentiation that show peculiar clinical and histomorphological features, with variable prognosis. In recent years, advances in knowledge regarding the pathophysiology and heterogeneous clinical presentation, as well as the availability of different diagnostic procedures for panNEN diagnosis and novel therapeutic options for patient clinical management, has led to the recognition of the need for an active multidisciplinary discussion for optimal patient care. Molecular imaging with positron emission tomography/computed tomography (PET/CT) has become indispensable for the management of panNENs. Several PET radiopharmaceuticals can be used to characterize either panNEN receptor expression or metabolism. The aim of this review is to offer an overview of all the currently used radiopharmaceuticals and of the new upcoming tracers for pancreatic neuroendocrine tumors (panNETs), and their clinical impact on therapy management. [68Ga]Ga-DOTA-peptide PET/CT (SSA-PET/CT) has high sensitivity, specificity, and accuracy and is recommended for the staging and restaging of any non-insulinoma well-differentiated panNEN cases to carry out detection of unknown primary tumor sites or early relapse and for evaluation of in vivo somatostatin receptors expression (SRE) to select patient candidates for peptide receptor radiometabolic treatment (PRRT) with 90Y or 177Lu and/or cold analogs. SSA-PET/CT also has a strong impact on clinical management, leading to a change in treatment in approximately a third of the cases. Its role for treatment response assessment is still under debate due to the lack of standardized criteria, even though some semiquantitative parameters seem to be able to predict response. [18F]FDG PET/CT generally shows low sensitivity in small growing and well-differentiated neuroendocrine tumors (NET; G1 and G2), while it is of utmost importance in the evaluation and management of high-grade NENs and also provides important prognostic information. When positive, [18F]FDG PET/CT impacts therapeutical management, indicating the need for a more aggressive treatment regime. Although FDG positivity does not exclude the patient from PRRT, several studies have demonstrated that it is certainly useful to predict response, even in this setting. The role of [18F]FDOPA for the study of panNET is limited by physiological uptake in the pancreas and is therefore not recommended. Moreover, it provides no information on SRE that has crucial clinical management relevance. Early acquisition of the abdomen and premedication with carbidopa may be useful to increase the accuracy, but further studies are needed to clarify its utility. GLP-1R agonists, such as exendin-4, are particularly useful for benign insulinoma detection, but their accuracy decreases in the case of malignant insulinomas. Being a whole-body imaging technique, exendin-PET/CT gives important preoperative information on tumor size and localization, which is fundamental for surgical planning as resection (enucleation of the lesion or partial pancreatic resection) is the only curative treatment. New upcoming tracers are under study, such as promising SSTR antagonists, which show a favorable biodistribution and higher tumor-to-background ratio that increases tumor detection, especially in the liver. [68Ga]pentixafor, an in vivo marker of CXCR4 expression associated with the behavior of more aggressive tumors, seems to only play a limited role in detecting well-differentiated NET since there is an inverse expression of SSTR2 and CXCR4 in G1 to G3 NETs with an elevation in CXCR4 and a decrease in SSTR2 expression with increasing grade. Other tracers, such as [68Ga]Ga-PSMA, [68Ga]Ga-DATA-TOC, [18F]SiTATE, and [18F]AlF-OC, are also under investigation.
Collapse
Affiliation(s)
- Diletta Calabrò
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Valentina Ambrosini
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
217
|
Lassmann M, Eberlein U, Tran-Gia J. Multicentre Trials on Standardised Quantitative Imaging and Dosimetry for Radionuclide Therapies. Clin Oncol (R Coll Radiol) 2020; 33:125-130. [PMID: 33277151 DOI: 10.1016/j.clon.2020.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/19/2020] [Accepted: 11/13/2020] [Indexed: 11/25/2022]
Abstract
The aim of this review is to summarise the efforts undertaken so far to compare or standardise quantitative imaging with gamma cameras across centres for multicentre trials in radionuclide therapies. Overall, 10 studies were identified, five of which were set up as a multicentre effort for standardising and comparing methods for quantitative imaging. One study used positron emission tomography imaging with 124I. In the remaining studies, measurements were carried out with planar imaging, single photon emission computed tomography/computed tomography (SPECT/CT) or a combination of both. Three studies used radioactive calibration sources that were traceable to national standards. Most of the studies were set up in the framework of multicentre clinical trials in an effort to obtain comparable quantification across sites. The use of state-of-the-art SPECT/CT systems and reconstructions has emerged as the method of choice for dosimetry in clinical trials for radionuclide therapies.
Collapse
Affiliation(s)
- M Lassmann
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany.
| | - U Eberlein
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany
| | - J Tran-Gia
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
218
|
Özdirik B, Amthauer H, Schatka I, Goretzki PE, Mogl MT, Fehrenbach U, Tacke F, Jann H, Roderburg C. A rare case of a patient with a high grade neuroendocrine tumor developing neutropenic sepsis after receiving PRRT combined with Capecitabine or Temozolomide: A case report. Mol Clin Oncol 2020; 14:20. [PMID: 33363730 PMCID: PMC7725216 DOI: 10.3892/mco.2020.2182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine tumors (NET) are rare and demonstrate variable clinical behavior depending on the degree of tumor differentiation. Patients with poorly differentiated tumors (NET G3) have a poor prognosis. Systemic treatment with cytotoxic chemotherapy is considered to be the treatment of choice. In patients that are refractory or intolerant to first-line therapy, experts recommend peptide receptor radionuclide therapy (PRRT) in tumors that express somatostatin receptors. Recently, combinations of PRRT and chemotherapy were tested in patients with NET. Available data have reported promising tumor control rates and an excellent toxicity profile in cases where PRRT had been administered with capecitabine/temozolomide, even when administered as salvage therapy. The current study reported an exceptional case of advanced NET G3 with severe toxicity upon receiving PRRT in combination with capecitabine/temozolomide as third line therapy. The patient developed a life-threatening neutropenic fever, fungal pneumonia and necrotizing mastitis 23 days after the first cycle of therapy was administered. However, the treatment led to a significant reduction in tumor size. A total of 5 months after treatment initiation, the patient was alive and in excellent clinical condition with sustained tumor response. In summary, the current study presented a rare case of high grade NET exhibiting an almost complete response to PRRT in combination capecitabine/temozolomide, despite facing unexpected severe toxicity.
Collapse
Affiliation(s)
- Burcin Özdirik
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Peter E Goretzki
- Department of Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Martina T Mogl
- Department of Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Uli Fehrenbach
- Diagnostic and Interventional Radiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Henning Jann
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| |
Collapse
|
219
|
D’Amico G, Uso TD. Liver Transplantation for Neuroendocrine Metastases. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
220
|
Grossrubatscher E, Fanciulli G, Pes L, Sesti F, Dolci C, de Cicco F, Colao A, Faggiano A. Advances in the Management of Medullary Thyroid Carcinoma: Focus on Peptide Receptor Radionuclide Therapy. J Clin Med 2020; 9:jcm9113507. [PMID: 33138305 PMCID: PMC7693147 DOI: 10.3390/jcm9113507] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Abstract
Effective treatment options in advanced/progressive/metastatic medullary thyroid carcinoma (MTC) are currently limited. As in other neuroendocrine neoplasms (NENs), peptide receptor radionuclide therapy (PRRT) has been used as a therapeutic option in MTC. To date, however, there are no published reviews dealing with PRRT approaches. We performed an in-depth narrative review on the studies published in this field and collected information on registered clinical trials related to this topic. We identified 19 published studies, collectively involving more than 200 patients with MTC, and four registered clinical trials. Most cases of MTC were treated with PRRT with somatostatin analogues (SSAs) radiolabelled with 90 yttrium (90Y) and 177 lutetium (177Lu). These radiopharmaceuticals show efficacy in the treatment of patients with MTC, with a favourable radiological response (stable disease, partial response or complete response) in more than 60% of cases, coupled with low toxicity. As MTC specifically also expresses cholecystokinin receptors (CCK2Rs), PRRT with this target has also been tried, and some randomised trials are ongoing. Overall, PRRT seems to have an effective role and might be considered in the therapeutic strategy of advanced/progressive/metastatic MTC.
Collapse
Affiliation(s)
- Erika Grossrubatscher
- Endocrine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
- Correspondence: (E.G.); (G.F.); Tel.: +39-026-444-3666 (E.G.); +39-079-264-4776 (G.F.)
| | - Giuseppe Fanciulli
- NET Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari-Endocrine Unit, AOU Sassari, 07100 Sassari, Italy
- Correspondence: (E.G.); (G.F.); Tel.: +39-026-444-3666 (E.G.); +39-079-264-4776 (G.F.)
| | - Luca Pes
- Endocrine Clinic, Policlinico Sassarese, 07100 Sassari, Italy;
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (A.F.)
| | - Carlotta Dolci
- Nuclear Medicine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Federica de Cicco
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, 80131 Naples, Italy; (F.d.C.); (A.C.)
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, 80131 Naples, Italy; (F.d.C.); (A.C.)
| | - Antongiulio Faggiano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (F.S.); (A.F.)
| | | |
Collapse
|
221
|
Kamaleshwaran KK, Suneelkumar M, Madhusairam R, Radhakrishnan EK, Arunpandiyan S, Arnold VJ. Whole-body and Single-Photon Emission Computed Tomography/Computed Tomography Postpeptide Receptor Alpha Radionuclide Therapy Images of Actinium 225-Tetraazacyclododecanetetraacetic Acid-Octreotide as a Primary Modality of Treatment in a Patient with Advanced Rectal Neuroendocrine Tumor with Metastases. Indian J Nucl Med 2020; 35:226-228. [PMID: 33082679 PMCID: PMC7537929 DOI: 10.4103/ijnm.ijnm_58_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 03/30/2020] [Accepted: 04/04/2020] [Indexed: 01/07/2023] Open
Abstract
Somatostatin receptor-targeted alpha radionuclide therapies have been introduced including actinium-225 labeled tetraazacyclododecanetetraacetic acid–octreotide (Ac-225 DOTATATE) for advanced metastatic neuroendocrine tumors (NET). Very limited data are available on the posttherapy imaging using Ac-225 DOTATATE therapy in metastatic NET. We present Ac-225 single-photon emission computed tomography/computed tomography images of a 72-year-old patient diagnosed as a case of advanced rectal NET treated primarily with Ac-225 DOTATATE.
Collapse
Affiliation(s)
- Koramadai Karuppusamy Kamaleshwaran
- Department of Nuclear Medicine, PET/CT and Radionuclide Therapy, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| | - Malipedda Suneelkumar
- Department of Nuclear Medicine, PET/CT and Radionuclide Therapy, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| | - Raghunathan Madhusairam
- Department of Radiation Oncology, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| | - Edathuruthy Kalarikal Radhakrishnan
- Department of Nuclear Medicine, PET/CT and Radionuclide Therapy, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| | - Soundararajan Arunpandiyan
- Department of Nuclear Medicine, PET/CT and Radionuclide Therapy, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| | - Vasumathi Jayaraj Arnold
- Department of Nuclear Medicine, PET/CT and Radionuclide Therapy, Comprehensive Cancer Care Center, Kovai Medical Center and Hospital Limited, Coimbatore, Tamil Nadu, India
| |
Collapse
|
222
|
Parghane RV, Mitra A, Bannore TU, Rakshit S, Banerjee S, Basu S. Initial clinical evaluation of indigenous 90Y-DOTATATE in sequential duo-PRRT approach ( 177Lu-DOTATATE and 90Y-DOTATATE) in neuroendocrine tumors with large bulky disease: Observation on tolerability, 90Y-DOTATATE post- PRRT imaging characteristics (bremsstrahlung and PETCT) and early adverse effects. World J Nucl Med 2020; 20:73-81. [PMID: 33850492 PMCID: PMC8034792 DOI: 10.4103/wjnm.wjnm_52_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 05/29/2020] [Accepted: 06/13/2020] [Indexed: 11/25/2022] Open
Abstract
177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) alone has lesser potential in the clinical setting of neuroendocrine tumor (NET) with large bulky disease and nonhomogeneous somatostatin receptors (SSTR) distribution, owing to lower energy (Eβmax 0.497 MeV) and a shorter particle penetration range (maximum 2–4 mm) of 177Lu. In large bulky NETs, 90Yttrium (90Y) has the theoretical advantages because of a longer beta particle penetration range (a maximum soft tissue penetration of 11 mm). Therefore, a combination of 177Lu and 90Y is a theoretically sound concept that can result in better response in metastatic NET with large-bulky lesion and non-homogeneous SSTR distribution. The aim of the study was to determine the feasibility of combining 90Y-DOTATATE with 177Lu-DOTATATE PRRT as sequential duo-PRRT in metastatic NET with (≥5 cm) including the post 90Y-DOTATATE-PRRT imaging and also to determine early toxicity of the duo-PRRT approach. A total of 9 patients received combination of 177Lu-DOTATATE with 90Y-DOTATATE (indigenously prepared and approved) through sequential duo-PRRT approach. These 9 NET patients were included and analyzed in this study. All 9 patients had undergone post-PRRT 90Y-DOTATATE imaging, including a whole-body planar bremsstrahlung imaging followed by regional single-photon emission computed tomography (SPECT)-computed tomography (CT) imaging and also a regional positron emission tomography–computed tomography imaging. Grading of 90Y-DOTATATE and 177Lu-DOTATATE uptake was done on post-PRRT imaging by both modalities. The size of the lesions ranged from 5.5 cm to 16 cm with average size of 10 cm before sequential duo-PRRT was decided. Sequential duo-PRRT was administered because of stable, unresponsive disease following 177Lu-DOTATATE in 5 patients (55.6%), progressive disease after 177Lu-DOTATATE in 2 patients (22.2%), and with neoadjuvant intent in 2 patients (22.2%). The total cumulative dose of 177Lu-DOTATATE before duo-pRRT ranged from 11.84 GBq to 37 GBq per patient and average administered dose of 27.21 GBq per patient in this study. Out of 9 patients, 8 patients received single cycle of 90Y-DOTATATE (ranging from 2.66 GBq to 3.4 GBq per patient with average administered dose of 3.12 GBq per patient). One patient received two cycles of 90Y-DOTATATE (total dose of 6.2 GBq). Out of 9 patients, 8 patients showed excellent tracer concentration in lesions on post-PRRT 90Y-DOTATATE imaging and the remaining 1 patient showed fairly adequate 90Y-DOTATATE tracer uptake in lesion on visual analysis. There was matched 90Y-DOTATATE uptake with 68Ga-DOTATATE and also with 177LuDOTATATE in all 9 patients. The sequential duo-PRRT was well tolerated by all patients. Two patients (22.2%) developed mild nausea, one patient (11.1%) developed transient mild-grade hemoglobin toxicity, and one patient (11.1%) developed mild-grade gastrointestinal symptoms (loose motion and abdominal pain). No nephrotoxicity, hepatotoxicity, and other hematological toxicity was observed. The combination of the indigenous 90Y-DOTATATE with 177Lu-DOTATATE PRRT in NET as sequential duo-PRRT was well tolerated, feasible and safe in stable, unresponsive/progressive disease following single isotope 177Lu-DOTATATE therapy and also in neoadjuvant PRRT setting with large bulky lesion (≥≥5cm). Post-PRRT 90Y-DOTATATE imaging showed excellent 90Y-DOTATATE uptake in nearly all NET patients. Mild-grade early adverse effects were easily manageable and controllable in this sequential duo-PRRT approach.
Collapse
Affiliation(s)
- Rahul V Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Arpit Mitra
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Trupti Upadhye Bannore
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sharmila Banerjee
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
223
|
Abstract
PURPOSE OF REVIEW Although the majority of pheochromocytoma and paraganglioma are benign, 15-17% develop metastatic disease, being present at the initial diagnosis in about 11-31% of cases. The natural course of metastasized disease is highly heterogeneous, with an overall 5-year survival rate varying between 40% and 85%. For individual patients, overall survival, progression-free survival, and clinical outcome are difficult to predict. Management of metastasized pheochromocytoma and paraganglioma is challenging. Currently available therapeutic options are surgical debulking, treatment with radiopharmaceuticals (I-MIBG, Y and Lu-DOTATATE), chemotherapy and targeted therapy. RECENT FINDINGS The pathogenesis of pheochromocytoma and paraganglioma (PPGL) is largely driven by genomic alterations in PPGL susceptibility genes related to three different clusters: altered pseudo-hypoxic signaling (cluster-1), altered MAP-kinase signaling (cluster-2) and altered Wnt signaling (cluster-3). Novel targeted therapies (tyrosine kinase inhibitors) and potential future therapeutic options, guided by improved knowledge about the oncogenic cluster 1-3 signaling pathways, will be discussed. SUMMARY Treatment of metastasized pheochromocytoma and paraganglioma remains challenging. Profiling of gene expression and methylation can serve as a powerful tool for characterizing disease clusters and for guiding targeted therapy to improve selectivity and efficacy. Current knowledge of signatures involved in molecular signaling, metabolism, and resistance mechanisms of PPGLs suggests that therapeutic regimens can be optimized to each molecular subtype.
Collapse
|
224
|
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
|
225
|
Müther M, Roll W, Brokinkel B, Zinnhardt B, Sporns PB, Seifert R, Schäfers M, Weckesser M, Stegger L, Stummer W, Rahbar K. Response assessment of somatostatin receptor targeted radioligand therapies for progressive intracranial meningioma. Nuklearmedizin 2020; 59:348-355. [PMID: 32691404 DOI: 10.1055/a-1200-0989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND In somatostatin receptor (SSTR) expressing progressive meningioma, peptide receptor radionuclide therapy (PRRT) has shown effect in small clinical series. However, standardized treatment and response assessment protocols are lacking. We present our experience on PPRT with 177Lu-DOTATATE in progressive meningioma with a special emphasis on state-of-the-art response assessment. METHODS Retrospective analysis on PRRT with 177Lu-DOTATATE from 2015 to 2019. Pre- and post-therapy imaging was performed using MRI and 68Ga-DOTATATE-PET for standard bidimensional and volumetric analyses, respectively, following novel RANO guidelines. RESULTS Seven patients with progressive intracranial meningioma (median age 73 years, interquartile range 60-76; 5 WHO II, 2 WHO I; 5 multifocal) received a median of 4 cycles 2 3 4 of PRRT with 177Lu-DOTATATE in eight-week intervals. Three patients did not undergo post-therapy 68Ga-DOTATATE-PET due to early symptomatic progression and subsequent cessation of PRRT. After completion of 4 PRRT cycles volumetric PET imaging showed stable disease in two of four patients. According to bidimensional MRI response assessment, only one patient was stable. Progression free survival at six months was 42.9 %. CONCLUSION In this heterogeneous collective of seven patients with progressive meningioma, 177Lu-DOTATATE therapies showed heterogeneous effectiveness. PET-based volumetric assessment should be used for response assessment in PRRT additionally to bidimensional imaging.
Collapse
Affiliation(s)
- Michael Müther
- Department of Neurosurgery, University Hospital Münster, Germany
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Germany
| | | | | | - Peter B Sporns
- Institute of Clinical Radiology, University Hospital Münster, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Münster, Germany
| | | | | | - Lars Stegger
- Department of Nuclear Medicine, University Hospital Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Germany
| |
Collapse
|
226
|
Gains JE, Moroz V, Aldridge MD, Wan S, Wheatley K, Laidler J, Peet C, Bomanji JB, Gaze MN. A phase IIa trial of molecular radiotherapy with 177-lutetium DOTATATE in children with primary refractory or relapsed high-risk neuroblastoma. Eur J Nucl Med Mol Imaging 2020; 47:2348-2357. [PMID: 32157433 DOI: 10.1007/s00259-020-04741-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE The objective of this phase IIa, open-label, single-centre, single-arm, two-stage clinical trial was to evaluate the safety and activity of 177-lutetium DOTATATE (LuDO) molecular radiotherapy in neuroblastoma. METHODS Children with relapsed or refractory metastatic high-risk neuroblastoma were treated with up to four courses of LuDO. The administered activity was 75 to 100 MBq kg-1 per course, spaced at 8- to 12-week intervals. Outcomes were assessed by the International Neuroblastoma Response Criteria (primary outcome), progression-free survival (PFS), and overall survival (OS). RESULTS The trial recruited 21 patients; eight received the planned four courses. There was dose-limiting haematologic toxicity in one case, but no other significant haematologic or renal toxicities. None of 14 evaluable patients had an objective response at 1 month after completion of treatment (Wilson 90% CI 0.0, 0.16; and 95% CI is 0.0, 0.22). The trial did not therefore proceed to the second stage. The median PFS was 2.96 months (95% CI 1.71, 7.66), and the median OS was 13.0 months (95% CI 2.99, 21.52). CONCLUSION In the absence of any objective responses, the use of LuDO as a single agent at the dose schedule used in this study is not recommended for the treatment of neuroblastoma. There are several reasons why this treatment schedule may not have resulted in objective responses, and as other studies do show benefit, the treatment should not be regarded as being of no value. Further trials designed to overcome this schedule's limitations are required. TRIAL REGISTRATION ISRCTN98918118; URL: https://www.isrctn.com/search?q=98918118.
Collapse
Affiliation(s)
- Jennifer E Gains
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
| | - Veronica Moroz
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Matthew D Aldridge
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Simon Wan
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Keith Wheatley
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jennifer Laidler
- Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Connie Peet
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK
| | - Jamshed B Bomanji
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK.
| |
Collapse
|
227
|
Basu S, Parghane RV, Kamaldeep, Chakrabarty S. Peptide Receptor Radionuclide Therapy of Neuroendocrine Tumors. Semin Nucl Med 2020; 50:447-464. [PMID: 32768008 DOI: 10.1053/j.semnuclmed.2020.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT), over the years, has evolved as an important modality in the therapeutic armamentarium of advanced, metastatic or inoperable, progressive Neuroendocrine Neoplasms (NENs). This review deliberates on the basic understanding and applied clinical aspects of PRRT in NENs, with special reference to (1) tumor biology and receptor characteristics, (2) molecular PET-CT imaging (in particular the invaluable role of dual-tracer PET with [68Ga]-DOTA-TATE/NOC and [18F]-FDG for exploring tumor biology in continuum and individualizing treatment decision making) and NEN theranostics, (3) relevant radiochemistry of different therapeutic radionuclides (both beta emitting 177Lu-DOTATATE and 90Y-DOTATATE and alpha emitting 225Ac-DOTATATE), and (4) related dosimetric considerations. Successful clinical management of the NENs would require multifactorial considerations, and all the aforementioned points pertaining to the disease process and available logistics are key considerations for state-of-the-art clinical practice and delivering personalized care in this group of patients. Emphasis has been placed on relatively intriguing areas such as (1) NET grade 3 of WHO 2017 classification (ie, Ki-67>20% but well-differentiation features), (2) "Neoadjuvant PRRT," (3) combining chemotherapy and PRRT, (4) 'Sandwich Chemo-PRRT', (5) duo-PRRT and tandem PRRT, (6) resistant functioning disease with nuances in clinical management and how one can advocate PRRT rationally in such clinical settings and individualize the management in a patient specific manner. Relevant clinical management issues related to some difficult case scenarios, which the Nuclear Medicine attending physician should be aware of to run an efficient clinical PRRT services, are described.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, India; Homi Bhabha National Institute, Mumbai, India.
| | - Rahul V Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Kamaldeep
- Homi Bhabha National Institute, Mumbai, India; Health Physics Division, Bhabha Atomic Research Centre Mumbai, India
| | - Sudipta Chakrabarty
- Homi Bhabha National Institute, Mumbai, India; Radiochemicals Section, Radiopharmaceuticals Division, Bhabha Atomic Research Centre Mumbai, India
| |
Collapse
|
228
|
Werner RA, Derlin T, Rowe SP, Bundschuh L, Sheikh GT, Pomper MG, Schulz S, Higuchi T, Buck AK, Bengel FM, Bundschuh RA, Lapa C. High Interobserver Agreement for the Standardized Reporting System SSTR-RADS 1.0 on Somatostatin Receptor PET/CT. J Nucl Med 2020; 62:514-520. [PMID: 32859702 DOI: 10.2967/jnumed.120.245464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/20/2020] [Indexed: 11/16/2022] Open
Abstract
Recently, a standardized framework system for interpreting somatostatin receptor (SSTR)-targeted PET/CT, termed the SSTR reporting and data system (RADS) 1.0, was introduced, providing reliable standards and criteria for SSTR-targeted imaging. We determined the interobserver reliability of SSTR-RADS for interpretation of 68Ga-DOTATOC PET/CT scans in a multicentric, randomized setting. Methods: A set of 51 randomized 68Ga-DOTATOC PET/CT scans was independently assessed by 4 masked readers with different levels of experience (2 experienced readers and 2 inexperienced readers) trained on the SSTR-RADS 1.0 criteria (based on a 5-point scale from 1 [definitively benign] to 5 [high certainty that neuroendocrine neoplasia is present]). For each scan, SSTR-RADS scores were assigned to a maximum of 5 target lesions (TLs). An overall scan impression based on SSTR-RADS was indicated, and interobserver agreement rates on a TL-based, on an organ-based, and on an overall SSTR-RADS score-based level were computed. The readers were also asked to decide whether peptide receptor radionuclide therapy (PRRT) should be considered on the basis of the assigned RADS scores. Results: Among the selected TLs, 153 were chosen by at least 2 readers (all 4 readers selected the same TLs in 58 of 153 [37.9%] instances). The interobserver agreement for SSTR-RADS scoring among identical TLs was good (intraclass correlation coefficient [ICC] ≥ 0.73 for 4, 3, and 2 identical TLs). For lymph node and liver lesions, excellent interobserver agreement rates were derived (ICC, 0.91 and 0.77, respectively). Moreover, the interobserver agreement for an overall scan impression based on SSTR-RADS was excellent (ICC, 0.88). The SSTR-RADS-based decision to use PRRT also demonstrated excellent agreement, with an ICC of 0.80. No significant differences between experienced and inexperienced readers for an overall scan impression and TL-based SSTR-RADS scoring were observed (P ≥ 0.18), thereby suggesting that SSTR-RADS seems to be readily applicable even for less experienced readers. Conclusion: SSTR-RADS-guided assessment demonstrated a high concordance rate, even among readers with different levels of experience, supporting the adoption of SSTR-RADS for trials, clinical routine, or outcome studies.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lena Bundschuh
- Department of Nuclear Medicine, University Medical Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Gabriel T Sheikh
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sebastian Schulz
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; and.,Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; and
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Medical Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
229
|
Basu S, Chakraborty S, Parghane RV, Kamaldeep, Ranade R, Thapa P, Asopa RV, Sonawane G, Nabar S, Shimpi H, Chandak A, Vimalnath KV, Ostwal V, Ramaswamy A, Bhandare M, Chaudhari V, Shrikhande SV, Sirohi B, Dash A, Banerjee S. One decade of 'Bench-to-Bedside' peptide receptor radionuclide therapy with indigenous [ 177Lu]Lu-DOTATATE obtained through 'Direct' neutron activation route: lessons learnt including practice evolution in an Indian setting. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2020; 10:178-211. [PMID: 32929396 PMCID: PMC7486551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/14/2020] [Indexed: 03/30/2024]
Abstract
The present treatise chronicles one decade of experience pertaining to clinical PRRT services in a large-volume tertiary cancer care centre in India delivering over 4,000 therapies, an exemplar of successful PRRT programme employing indigenous 177Lutetium production and resources. For the purpose of systematic discussion, we have sub-divided the communication into 3 specific parts: (a) Radiopharmaceutical aspects that describes 177Lutetium production through 'Direct' Neutron Activation Route and the subsequent radiolabeling procedures, (b) The specific clinical nuances and finer learning points (apart from the routine standard procedure) based upon clinical experience and how it has undergone practice evolution in our setting and (c) Dosimetry results with this indigenous product and radiation safety/health physics aspects involved in PRRT services. Initiated in 2010 at our centre, the PRRT programme is a perfect example of affordable quality health care delivery, with indigenous production of the radionuclide (177Lu) in the reactor and subsequent radiolabeling of the radiopharmaceutical ([177Lu]Lu-DOTATATE) at the hospital radiopharmacy unit of the centre, which enabled catering to the needs of a large number of patients of progressive, metastatic and advanced Neuroendocrine Neoplasms (NENs) and related malignancies.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Sudipta Chakraborty
- Homi Bhabha National InstituteMumbai, India
- Radiopharmaceuticals Division, BARCMumbai, India
| | - Rahul V Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Kamaldeep
- Homi Bhabha National InstituteMumbai, India
- Health Physics Division, Bhabha Atomic Research CentreMumbai, India
| | - Rohit Ranade
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Pradeep Thapa
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Ramesh V Asopa
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Geeta Sonawane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Swapna Nabar
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Hemant Shimpi
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - Ashok Chandak
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| | - KV Vimalnath
- Radiopharmaceuticals Division, BARCMumbai, India
| | - Vikas Ostwal
- Homi Bhabha National InstituteMumbai, India
- Department of Medical Oncology, Tata Memorial CentreMumbai, Maharashtra, India
| | - Anant Ramaswamy
- Homi Bhabha National InstituteMumbai, India
- Department of Medical Oncology, Tata Memorial CentreMumbai, Maharashtra, India
| | - Manish Bhandare
- Homi Bhabha National InstituteMumbai, India
- Department of Surgical Oncology, Gastrointestinal and Hepato-Pancreato-Biliary Service, Tata Memorial HospitalMumbai, India
| | - Vikram Chaudhari
- Homi Bhabha National InstituteMumbai, India
- Department of Surgical Oncology, Gastrointestinal and Hepato-Pancreato-Biliary Service, Tata Memorial HospitalMumbai, India
| | - Shailesh V Shrikhande
- Homi Bhabha National InstituteMumbai, India
- Department of Surgical Oncology, Gastrointestinal and Hepato-Pancreato-Biliary Service, Tata Memorial HospitalMumbai, India
| | - Bhawna Sirohi
- Department of Medical Oncology, Tata Memorial CentreMumbai, Maharashtra, India
- Apollo Proton Cancer CentreChennai, India
| | - Ashutosh Dash
- Homi Bhabha National InstituteMumbai, India
- Radiopharmaceuticals Division, BARCMumbai, India
| | - Sharmila Banerjee
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital AnnexeParel, Mumbai, India
- Homi Bhabha National InstituteMumbai, India
| |
Collapse
|
230
|
Sulieman A, Mayhoub FH, Salah H, Al-Mohammed HI, Alkhorayef M, Moftah B, Al Rowaily M, Bradley DA. Occupational and ambient radiation exposures from Lu-177 DOTATATE during targeted therapy. Appl Radiat Isot 2020; 164:109240. [PMID: 32819499 DOI: 10.1016/j.apradiso.2020.109240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/22/2020] [Accepted: 05/19/2020] [Indexed: 11/16/2022]
Abstract
Lutetium-177 (DOTATATE) (177Lu; T1/2 6.7 days), a labelled β- and Auger-electron emitter, is widely used in treatment of neuroendocrine tumours. During performance of the procedure, staff and other patients can potentially receive significant doses in interception of the gamma emissions [113 keV (6.4%) and 208 keV (11%)] that are associated with the particle decays. While radiation protection and safety assessment are required in seeking to ensure practices comply with international guidelines, only limited published studies are available. The objectives of present study are to evaluate patient and occupational exposures, measuring ambient doses and estimating the radiation risk. The results, obtained from studies carried out in Riyadh over an 11 month period, at King Faisal Specialist Hospital and Research Center, concerned a total of 33 177Lu therapy patients. Patient exposures were estimated using a calibrated Victoreen 451P survey meter (Fluke Biomedical), for separations of 30 cm, 100 cm and 300 cm, also behind a bed shield that was used during hospitalization of the therapy patients. Occupational and ambient doses were also measured through use of calibrated thermoluminescent dosimeters and an automatic TLD reader (Harshaw 6600). The mean and range of administered activity (in MBq)) was 7115.2 ± 917.2 (4329-7955). The ambient dose at corridors outside of therapy isolation rooms was 1.2 mSv over the 11 month period, that at the nursing station was below the limit of detection and annual occupational doses were below the annual dose limit of 20 mSv. Special concern needs to be paid to comforters (carers) and family members during the early stage of radioisotope administration.
Collapse
Affiliation(s)
- A Sulieman
- Prince Sattam Bin Abdulaziz University, College of Applied Medical Sciences, Radiology and Medical Imaging Department, P.O.Box 422, Alkharj, 11942, Saudi Arabia.
| | - Fareed H Mayhoub
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Hassan Salah
- INAYA Medical Collage, Nuclear Medicine Department, Riyadh, 13541, Saudi Arabia; College of Medical Radiologic Science, Sudan University of Science and Technology, P.O.Box 1908, Khartoum, 11111, Sudan
| | - H I Al-Mohammed
- Department of Radiological Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, P.O Box 84428, Riyadh, 11671, Saudi Arabia
| | - M Alkhorayef
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, P.O Box 10219, Riyadh, 11433, Saudi Arabia; Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford, Surrey GU2 7XH, UK
| | - B Moftah
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - M Al Rowaily
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - D A Bradley
- Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford, Surrey GU2 7XH, UK; Centre for Biomedical Physics, School of Healthcare and Medical Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
231
|
Maughan NM, Kim H, Hao Y, Unangst S, Roach MC, Garcia-Ramirez JL, Amurao M, Luechtefeld D, Abdin K, Altman MB, Banks A, Riepe M, Bovard E, Jenkins S, Zoberi JE. Initial experience and lessons learned with implementing Lutetium-177-dotatate radiopharmaceutical therapy in a radiation oncology-based program. Brachytherapy 2020; 20:237-247. [PMID: 32819853 DOI: 10.1016/j.brachy.2020.07.004] [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] [Received: 12/31/2019] [Revised: 06/05/2020] [Accepted: 07/02/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To assist radiation oncology centers in implementing Lutetium-177-dotatate (177Lu) radiopharmaceutical therapy for midgut neuroendocrine tumors. Here we describe our workflow and how it was revised based on our initial experience on an expanded access protocol (EAP). METHODS A treatment team/area was identified. An IV-pump-based infusion technique was implemented. Exposure-based techniques were implemented to determine completion of administration, administered activity, and patient releasability. Acute toxicities were assessed at each fraction. A workflow failure modes and effects analysis (FMEA) was performed. RESULTS A total of 22 patients were treated: 11 patients during EAP (36 administrations) and 11 patients after EAP (44 administrations). Mean 177Lu infusion time was 37 min (range 26-65 min). Mean administered activity was 97% (range 90-99%). Mean patient exposures at 1 m were 1.9 mR/h (range 1.0-4.1 mR/h) post-177Lu and 0.9 mR/h (range 0.4-1.8 mR/h) at discharge, rendering patients releasable with instructions. Treatment area was decontaminated and released same day. All patients in the EAP experienced nausea, and nearly half experienced emesis despite premedication with antiemetics. Peripheral IV-line complications occurred in six treatments (16.7%), halting administration in 2 cases (5.6%). We transitioned to peripherally inserted central catheter (PICC)-lines and revised amino acid formulary after the EAP. The second cohort of 11 patients after EAP were analyzed for PICC-line complications and acute toxicity. Nausea and emesis rates decreased (nausea G1+ 61%-27%; emesis G1+ 23%-7%), and no PICC complications were observed. FMEA revealed that a failure in amino acid preparation was the highest risk. CONCLUSION 177Lu-dotatate can be administered safely in an outpatient radiation oncology department.
Collapse
Affiliation(s)
- Nichole M Maughan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO.
| | - Hyun Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Yao Hao
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | | | - Michael C Roach
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Jose L Garcia-Ramirez
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Max Amurao
- Radiation Safety Division, EH&S Department, Washington University in St. Louis, St Louis, MO
| | - David Luechtefeld
- Radiation Safety Division, EH&S Department, Washington University in St. Louis, St Louis, MO
| | - Kinda Abdin
- Radiation Safety Division, EH&S Department, Washington University in St. Louis, St Louis, MO
| | - Michael B Altman
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Alexis Banks
- Department of Radiation Oncology, Barnes Jewish Hospital, St. Louis, MO
| | - Mary Riepe
- Department of Radiation Oncology, Barnes Jewish Hospital, St. Louis, MO
| | - Erica Bovard
- Department of Radiation Oncology, Barnes Jewish Hospital, St. Louis, MO
| | - Samantha Jenkins
- Department of Radiation Oncology, Barnes Jewish Hospital, St. Louis, MO
| | - Jacqueline E Zoberi
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
232
|
Huizing DMV, Aalbersberg EA, Versleijen MWJ, Tesselaar MET, Walraven I, Lahaye MJ, de Wit-van der Veen BJ, Stokkel MPM. Early response assessment and prediction of overall survival after peptide receptor radionuclide therapy. Cancer Imaging 2020; 20:57. [PMID: 32778165 PMCID: PMC7418334 DOI: 10.1186/s40644-020-00335-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/29/2020] [Indexed: 01/19/2023] Open
Abstract
Background Response after peptide receptor radionuclide therapy (PRRT) can be evaluated using anatomical imaging (CT/MRI), somatostatin receptor imaging ([68Ga]Ga-DOTA-TATE PET/CT), and serum Chromogranin-A (CgA). The aim of this retrospective study is to assess the role of these response evaluation methods and their predictive value for overall survival (OS). Methods Imaging and CgA levels were acquired prior to start of PRRT, and 3 and 9 months after completion. Tumour size was measured on anatomical imaging and response was categorized according to RECIST 1.1 and Choi criteria. [68Ga]Ga-DOTA-TATE uptake was quantified in both target lesions depicted on anatomical imaging and separately identified PET target lesions, which were either followed over time or newly identified on each scan with PERCIST-based criteria. Response evaluation methods were compared with Cox regression analyses and Log Rank tests for association with OS. Results A total of 44 patients were included, with median follow-up of 31 months (IQR 26–36 months) and median OS of 39 months (IQR 32mo-not reached)d. Progressive disease after 9 months (according to RECIST 1.1) was significantly associated with worse OS compared to stable disease [HR 9.04 (95% CI 2.10–38.85)], however not compared to patients with partial response. According to Choi criteria, progressive disease was also significantly associated with worse OS compared to stable disease [HR 6.10 (95% CI 1.38–27.05)] and compared to patients with partial response [HR 22.66 (95% CI 2.33–219.99)]. In some patients, new lesions were detected earlier with [68Ga]Ga-DOTA-TATE PET/CT than with anatomical imaging. After 3 months, new lesions on [68Ga]Ga-DOTA-TATE PET/CT which were not visible on anatomical imaging, were detected in 4/41 (10%) patients and in another 3/27 (11%) patients after 9 months. However, no associations between change in uptake on 68Ga-DOTA-TATE PET/CT or serum CgA measurements and OS was observed. Conclusions Progression on anatomical imaging performed 9 months after PRRT is associated with worse OS compared to stable disease or partial response. Although new lesions were detected earlier with [68Ga]Ga-DOTA-TATE PET/CT than with anatomical imaging, [68Ga]Ga-DOTA-TATE uptake, and serum CgA after PRRT were not predictive for OS in this cohort with limited number of patients and follow-up time.
Collapse
Affiliation(s)
- Daphne M V Huizing
- Department of Nuclear Medicine, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands.
| | - Else A Aalbersberg
- Department of Nuclear Medicine, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| | - Michelle W J Versleijen
- Department of Nuclear Medicine, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| | - Margot E T Tesselaar
- Department of Medical Oncology, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| | - Iris Walraven
- Department of Radiotherapy, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| | - Max J Lahaye
- Department of Radiology, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| | | | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, ENETS Center of Excellence, Amsterdam, The Netherlands
| |
Collapse
|
233
|
Comparison of different calculation techniques for absorbed dose assessment in patient specific peptide receptor radionuclide therapy. PLoS One 2020; 15:e0236466. [PMID: 32764764 PMCID: PMC7413508 DOI: 10.1371/journal.pone.0236466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/06/2020] [Indexed: 11/19/2022] Open
Abstract
Aim The present work concerns the comparison of the performances of three systems for dosimetry in RPT that use different techniques for absorbed dose calculation (organ-level dosimetry, voxel-level dose kernel convolution and Monte Carlo simulations). The aim was to assess the importance of the choice of the most adequate calculation modality, providing recommendations about the choice of the computation tool. Methods The performances were evaluated both on phantoms and patients in a multi-level approach. Different phantoms filled with a 177Lu-radioactive solution were used: a homogeneous cylindrical phantom, a phantom with organ-shaped inserts and two cylindrical phantoms with inserts different for shape and volume. A total of 70 patients with NETs treated by PRRT with 177Lu-DOTATOC were retrospectively analysed. Results The comparisons were performed mainly between the mean values of the absorbed dose in the regions of interest. A general better agreement was obtained between Dose kernel convolution and Monte Carlo simulations results rather than between either of these two and organ-level dosimetry, both for phantoms and patients. Phantoms measurements also showed the discrepancies mainly depend on the geometry of the inserts (e.g. shape and volume). For patients, differences were more pronounced than phantoms and higher inter/intra patient variability was observed. Conclusion This study suggests that voxel-level techniques for dosimetry calculation are potentially more accurate and personalized than organ-level methods. In particular, a voxel-convolution method provides good results in a short time of calculation, while Monte Carlo based computation should be conducted with very fast calculation systems for a possible use in clinics, despite its intrinsic higher accuracy. Attention to the calculation modality is recommended in case of clinical regions of interest with irregular shape and far from spherical geometry, in which Monte Carlo seems to be more accurate than voxel-convolution methods.
Collapse
|
234
|
Roll W, Müther M, Sporns PB, Zinnhardt B, Suero Molina E, Seifert R, Schäfers M, Weckesser M, Stegger L, Beule AG, Stummer W, Rahbar K. Somatostatin Receptor-Targeted Radioligand Therapy in Head and Neck Paraganglioma. World Neurosurg 2020; 143:e391-e399. [PMID: 32745642 DOI: 10.1016/j.wneu.2020.07.165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Surgical resection is the therapy of choice in head and neck paraganglioma but is associated with considerable morbidity. For treatment of inoperable or progressive disease, less aggressive adjuvant options are warranted. This study assessed effectiveness and safety of peptide receptor radionuclide therapy (PRRT) with lutetium-177-DOTATATE for head and neck paraganglioma with emphasis on response assessment. METHODS A retrospective analysis of 7 patients with head and neck paraganglioma treated with PPRT between May 2014 and October 2016 was performed. Three patients had jugulotympanic paraganglioma, 3 patients had carotid body tumors, and 1 patient had a combination of both. Patients underwent PRRT after discussion in the local tumor board regarding progressive disease, inoperability, or lack of other adjuvant options. All patients underwent 3-5 cycles of PRRT. Treatment response was evaluated by gallium-68-DOTATATE positron emission tomography/computed tomography and contrast-enhanced computed tomography or magnetic resonance imaging. Outcome measures were two-dimensional tumor diameters and total tumor volumes. RESULTS Median patient age was 60 years (interquartile range: 14-84 years). All patients had stable disease at posttherapy assessment. Decreasing tumor volumes were found in 4 patients. Clinical symptoms improved in 2 patients. No progression or adverse events occurred during a median follow-up of 39 months (interquartile range: 35-47 months). CONCLUSIONS Somatostatin receptor-targeted therapy using lutetium-177-DOTATATE shows promising effectiveness with a high safety profile. Patients in whom surgical morbidity outweighs oncologic benefit should be informed about PRRT as a treatment option.
Collapse
Affiliation(s)
- Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Michael Müther
- Department of Neurosurgery, University Hospital Münster, Münster, Germany.
| | - Peter B Sporns
- Institute of Clinical Radiology, University Hospital Münster, Münster, Germany
| | - Bastian Zinnhardt
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; European Institute of Molecular Imaging, University of Münster, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; European Institute of Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany; European Institute of Molecular Imaging, University of Münster, Münster, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Lars Stegger
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Achim G Beule
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Münster, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
235
|
Positron Emission Tomography-Based Response to Target and Immunotherapies in Oncology. ACTA ACUST UNITED AC 2020; 56:medicina56080373. [PMID: 32722205 PMCID: PMC7466359 DOI: 10.3390/medicina56080373] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/11/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) is a promising tool to support the evaluation of response to either target therapies or immunotherapy with immune checkpoint inhibitors both in clinical trials and, in selected patients, at the single patient's level. The present review aims to discuss available evidence related to the use of [18F]FDG PET (Positron Emission Tomography) to evaluate the response to target therapies and immune checkpoint inhibitors. Criteria proposed for the standardization of the definition of the PET-based response and complementary value with respect to morphological imaging are commented on. The use of PET-based assessment of the response through metabolic pathways other than glucose metabolism is also relevant in the framework of personalized cancer treatment. A brief discussion of the preliminary evidence for the use of non-FDG PET tracers in the evaluation of the response to new therapies is also provided.
Collapse
|
236
|
del Olmo-García MI, Muros MA, López-de-la-Torre M, Agudelo M, Bello P, Soriano JM, Merino-Torres JF. Prevention and Management of Hormonal Crisis during Theragnosis with LU-DOTA-TATE in Neuroendocrine Tumors. A Systematic Review and Approach Proposal. J Clin Med 2020; 9:E2203. [PMID: 32664679 PMCID: PMC7408760 DOI: 10.3390/jcm9072203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroendocrine tumors (NETs) frequently overexpress somatostatin receptors (SSTR) on their cell surface. The first-line pharmacological treatment for inoperable metastatic functioning well-differentiated NETs are somatostatin analogs. On second line, Lu-DOTA-TATE (177Lu-DOTA0 Tyr 3 octreotate) has shown stabilization of the disease and an increase in progression free survival, as well as effectiveness in controlling symptoms and increasing quality of life. The management of functional NETs before and during LU-DOTA-TATE treatment is specially challenging, as several complications such as severe carcinoid and catecholamine crisis have been described. The aim of this review is to establish practical guidance for the management and prevention of the most common hormonal crises during radionuclide treatment with Lu-DOTA-TATE: carcinoid syndrome (CS) and catecholamine hypersecretion, as well as to provide a brief commentary on other infrequent metabolic complications. To establish a practical approach, a systematic review was performed. This systematic review was developed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement and conducted using MEDLINE (accessed from PubMed), Google Scholar and ClinicalTrials.gov. Literature searches found 449 citations, and finally nine were considered for this systematic review.
Collapse
Affiliation(s)
| | - Maria Angustias Muros
- Nuclear Medicine Department, University Hospital Virgen de las Nieves, 18014 Granada, Spain;
| | - Martín López-de-la-Torre
- Endocrinology and Nutrition Department, University Hospital Virgen de las Nieves, 18014 Granada, Spain;
| | - Marc Agudelo
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, 46026 Valencia, Spain; (M.A.); (P.B.)
| | - Pilar Bello
- Nuclear Medicine Department, University and Polytechnic Hospital La Fe, 46026 Valencia, Spain; (M.A.); (P.B.)
| | - Jose M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute La Fe, 46026 Valencia, Spain
| | - Juan-Francisco Merino-Torres
- Endocrinology and Nutrition Department, University and Polytechnic Hospital La Fe, 46026 Valencia, Spain;
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute La Fe, 46026 Valencia, Spain
| |
Collapse
|
237
|
Jiménez-Franco LD, Glatting G, Prasad V, Weber WA, Beer AJ, Kletting P. Effect of Tumor Perfusion and Receptor Density on Tumor Control Probability in 177Lu-DOTATATE Therapy: An In Silico Analysis for Standard and Optimized Treatment. J Nucl Med 2020; 62:92-98. [PMID: 32646878 DOI: 10.2967/jnumed.120.245068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/30/2020] [Indexed: 01/02/2023] Open
Abstract
The aim of this work was to determine a minimal tumor perfusion and receptor density for 177Lu-DOTATATE therapy using physiologically based pharmacokinetic (PBPK) modeling considering, first, a desired tumor control probability (TCP) of 99% and, second, a maximal tolerated biologically effective dose (BEDmax) for organs at risk (OARs) in the treatment of neuroendocrine tumors and meningioma. Methods: A recently developed PBPK model was used. Nine virtual patients (i.e., individualized PBPK models) were used to perform simulations of pharmacokinetics for different combinations of perfusion (0.001-0.1 mL/g/min) and receptor density (1-100 nmol/L). The TCP for each combination was determined for 3 different treatment strategies: a standard treatment (4 cycles of 7.4 GBq and 105 nmol), a treatment maximizing the number of cycles based on BEDmax for red marrow and kidneys, and a treatment having 4 cycles with optimized ligand amount and activity. The red marrow and the kidneys (BEDmax of 2 Gy15 and 40 Gy2.5, respectively) were assumed to be OARs. Additionally, the influence of varying glomerular filtration rates, kidney somatostatin receptor densities, tumor volumes, and release rates was investigated. Results: To achieve a TCP of at least 99% in the standard treatment, a minimal tumor perfusion of 0.036 ± 0.023 mL/g/min and receptor density of 34 ± 20 nmol/L were determined for the 9 virtual patients. With optimization of the number of cycles, the minimum values for perfusion and receptor density were considerably lower, at 0.022 ± 0.012 mL/g/min and 21 ± 11 nmol/L, respectively. However, even better results (perfusion, 0.018 ± 0.009 mL/g/min; receptor density, 18 ± 10 nmol/L) were obtained for strategy 3. The release rate of 177Lu (or labeled metabolites) from tumor cells had the strongest effect on the minimal perfusion and receptor density for standard and optimized treatments. Conclusion: PBPK modeling and simulations represent an elegant approach to individually determine the minimal tumor perfusion and minimal receptor density required to achieve an adequate TCP. This computational method can be used in the radiopharmaceutical development process for ligand and target selection for specific types of tumors. In addition, this method could be used to optimize clinical trials.
Collapse
Affiliation(s)
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany.,Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany; and
| | - Vikas Prasad
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Peter Kletting
- Department of Nuclear Medicine, Ulm University, Ulm, Germany .,Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany; and
| |
Collapse
|
238
|
Lim LE, Chan DL, Thomas D, Du Y, Tincknell G, Kuchel A, Davis A, Bailey DL, Pavlakis N, Cehic G, Macdonald W, Wyld D, Segelov E. Australian experience of peptide receptor radionuclide therapy in lung neuroendocrine tumours. Oncotarget 2020; 11:2636-2646. [PMID: 32676165 PMCID: PMC7343632 DOI: 10.18632/oncotarget.27659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/15/2020] [Indexed: 01/29/2023] Open
Abstract
Background: Peptide receptor radionuclide therapy (PRRT) is an approved treatment modality for gastroenteropancreatic neuroendocrine tumours (GEP NETs), Although Phase III randomised clinical trial data is not available for NETs of other site of origin, in practice, PRRT is used more widely in clinical practice, based on its mechanism of targeting the somatostatin receptor. Use of PRRT for lung (bronchial) NET, specifically typical and atypical carcinoid (TC, AC), has been reported only in small retrospective case series. This multicentre study adds to the evidence regarding utility of PRRT for lung NETs. Materials and Methods: A retrospective chart review of patients with TC and AC who received 177Lu-dotatate PRRT between January 2002 and June 2019 in six hospitals across Australia was undertaken. Data regarding demographics, efficacy and toxicity was evaluated at each site by the treating clinician. Results: Forty-eight patients (32 AC, 15 TC, 1 unclassified) received a median of four 177Lu-dotatate treatments. There was a median of one prior line of systemic treatment (range: 0–3). The response rate to 177Lu-dotatate was 33%, with a median overall survival of 49 months (range of 3–91), at a median follow up of 33 months. This compares favourably with GEP NET. Overall toxicity was recorded as modest. Conclusions: 177Lu-dotatate PRRT in patients with lung NETs is used in real world practice, where it appears well-tolerated with some efficacy. Further evidence could be obtained through a global prospective clinical or registry trial.
Collapse
Affiliation(s)
| | - David L Chan
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia.,Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, University of Sydney, Sydney, Australia
| | - David Thomas
- Department of Medical Oncology, St. George Hospital, Sydney, Australia
| | - Yang Du
- Department of Nuclear Medicine, The Queen Elizabeth Hospital, Adelaide, Australia
| | - Gary Tincknell
- Department of Medical Oncology, St. George Hospital, Sydney, Australia
| | - Anna Kuchel
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Alexander Davis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Dale L Bailey
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, Sydney, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, Sydney, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Gabrielle Cehic
- Department of Nuclear Medicine, The Queen Elizabeth Hospital, Adelaide, Australia.,University of South Australia, Adelaide, Australia
| | - William Macdonald
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, Australia
| | - David Wyld
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Eva Segelov
- Department of Medical Oncology, Monash Health, Melbourne, Australia.,Faculty of Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
239
|
Tuncel M, Kılıçkap S, Süslü N. Clinical impact of 68Ga-DOTATATE PET-CT imaging in patients with medullary thyroid cancer. Ann Nucl Med 2020; 34:663-674. [PMID: 32602032 DOI: 10.1007/s12149-020-01494-3] [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] [Received: 05/20/2020] [Accepted: 06/17/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Medullary thyroid cancer (MTC) arises from neuroendocrine C cells of the thyroid. There is no single diagnostic imaging method that can reveal all MTC recurrences or metastases. 68Ga-DOTATATE is an alternative PET radiotracer that showed acceptable efficacy in the detection of MTC. In this study, we aimed to reveal the clinical efficacy and impact of this radiotracer on the management of patients with MTC. METHODS The 68Ga-DOTATATE PET-CT records of 38 patients with confirmed MTC were included in the study. The demographic data, clinical indication for the scan, previous therapies, and tumor marker levels were recorded. The site and SUVmax of the lesions were also noted. A consensus was reached on the additional value of 68Ga-DOTATATE PET-CT, and sites with discordant results on conventional imaging (CI). Finally, changes in management after the scan were evaluated. RESULTS 68Ga-DOTATATE PET-CT outperformed CI in 14/38 (37%) patients. In these 14 patients, metastatic lymph nodes were detected in 8, bone metastases in 4, and both bone and lymph nodes metastases in 2 patients. In 16/38 (42%) patients, 68Ga-DOTATATE PET-CT performed equally well as CI. In 5/38 (13%) patients, CI outperformed PET-CT. Most of the patients (4/5) in this group had hepatic metastases. 68Ga-DOTATATE PET-CT positivity was also correlated with tumor marker expression [median calcitonin; PET-positive: 743 ± 5439 vs PET-negative: 45 ± 17 (p:0.012), median CEA; PET-positive: 41 ± 162 vs PET-negative: 2.6 ± 1.4 (p:0.015)]. 68Ga-DOTATATE PET-CT changed the clinical management of 13/38 (34%) patients. The information provided by PET-CT resulted in neck surgery in 5/13 patients, external radiotherapy in 3/13 and both in one patient. Four of these thirteen patients were found to be eligible for peptide receptor radionuclide therapy. CONCLUSION 68 Ga-DOTATATE is an essential part of the work-up for patients with MTC. This modality outperformed CI in 14/38 (37%) patients and changed the clinical management in 13/38 (34%) patients. Prospective randomized studies with image-guided therapy decisions are needed to further reveal the impact of PET imaging in patients with MTC.
Collapse
Affiliation(s)
- Murat Tuncel
- Department of Nuclear Medicine, Faculty of Medicine, Hacettepe University, Sıhhiye, 06100, Ankara, Turkey.
| | - Saadettin Kılıçkap
- Department of Medical Oncology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Nilda Süslü
- Department of Head and Neck Surgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
240
|
Arveschoug AK, Bekker AC, Iversen P, Bluhme H, Villadsen GE, Staanum PF. Extravasation of [ 177Lu]Lu-DOTATOC: case report and discussion. EJNMMI Res 2020; 10:68. [PMID: 32577838 PMCID: PMC7311613 DOI: 10.1186/s13550-020-00658-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/09/2020] [Indexed: 12/02/2022] Open
Abstract
Background In the case of extravasation of radioactive drugs used in peptide-receptor radionuclide therapy of neuroendocrine tumors, or in radionuclide therapy in general, rapid action is important to reduce or avoid complications. The literature on extravasation of drugs for radionuclide therapy is sparse. Based on the present case, we discuss handling and consequences of extravasation. Further, we demonstrate that dosimetry can aid in judging if the treatment of neuroendocrine tumors is satisfactory even after extravasation. Case presentation A case of extravasation of [177Lu]Lu-DOTATOC with a treatment strategy involving exercise and elevation of the affected arm and application of a compression bandage and heating is reported. Redistribution of the drug is verified and quantified by whole-body imaging and quantitative SPECT/CT and measurements of the dose rate at contact with the injection site. [177Lu]Lu-DOTATOC was redistributed to tumors and organs within 1 day. The patient did not report any discomfort during or after hospitalization, and no side effects related to extravasation were observed. Quantitative SPECT/CT scans at the subsequent treatment cycle of the same patient were analyzed for a comparison between the treatments. Dosimetry showed the treatments were similar with respect to the kidney and tumor absorbed doses. The radiation dose to the epidermal basal layer near the injection site was estimated and found to be consistent with the lack of side effects. Conclusions The treatment of extravasation was successful, and the redistribution of the drug can be easily verified through measurement of the dose rate at contact with the skin. From the results of dosimetry, it was assessed that no change of the treatment course was necessary to compensate for a possibly incomplete treatment as a result of the extravasation.
Collapse
Affiliation(s)
- Anne Kirstine Arveschoug
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Anne Charlotte Bekker
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Peter Iversen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Henrik Bluhme
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark
| | - Gerda Elisabeth Villadsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Peter Frøhlich Staanum
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200, Aarhus N, Denmark.
| |
Collapse
|
241
|
Pediatric applications of Dotatate: early diagnostic and therapeutic experience. Pediatr Radiol 2020; 50:882-897. [PMID: 32495176 DOI: 10.1007/s00247-020-04688-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/17/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022]
Abstract
In recent years, new somatostatin receptor agents (SSTRs) have become available for diagnostic imaging and therapy in neuroendocrine tumors. The novel SSTR ligand DOTA-DPhel-Tyr3-octreotate (Dotatate) in particular can be linked with 68Gallium for diagnostic imaging purposes, and with the β-emitter 177Lutetium for radiotherapy in the setting of neuroendocrine tumors. Dotatate imaging offers distinct advantages in the evaluation of neuroendocrine tumors compared to standard techniques, including greater target-to-background ratio and lesion conspicuity, high sensitivity/specificity, improved spatial resolution with positron emission tomography (PET)/CT or PET/MR, and decreased radiation exposure. Although currently off-label in pediatrics, Dotatate theranostics in children are being explored, most notably in the setting of neuroblastoma and hereditary neuroendocrine syndromes. This article provides a multicenter case series of Dotatate imaging and therapy in pediatric patients in order to highlight the spectrum of potential clinical applications.
Collapse
|
242
|
Castillo Seoane D, de Saint-Hubert M, Crabbe M, Struelens L, Koole M. Targeted alpha therapy: a critical review of translational dosimetry research with emphasis on actinium-225. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:265-277. [PMID: 32441067 DOI: 10.23736/s1824-4785.20.03266-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This review provides a general overview of the current achievements and challenges in translational dosimetry for targeted alpha therapy (TAT). The concept of targeted radionuclide therapy (TRNT) is described with an overview of its clinical applicability and the added value of TAT is discussed. For TAT, we focused on actinium-225 (225Ac) as an example for alpha particle emitting radionuclides and their features, such as limited range within tissue and high linear energy transfer, which make alpha particle emissions more effective in targeted killing of tumour cells compared to beta radiation. Starting with the state-of-the-art dosimetry for TRNT and TAT, we then describe the challenges that still need to be met in order to move to a personalized dosimetry approach for TAT. Specifically for 225Ac, we discuss the recoiled daughter effect which may provoke significant damage to healthy tissue or organs and should be considered. Next, a broad overview is given of the pre-clinical research on 225Ac-TAT with an extensive description of tools which are only available in a pre-clinical setting and their added value. In addition, we review the preclinical biodistribution and dosimetry studies that have been performed on TAT-agents and more specifically of 225Ac and its multiple progeny, and describe their potential role to better characterize the pharmacokinetic (PK) profile of TAT-agents and to optimize the use of theranostic approaches for dosimetry. Finally, we discuss the support pre-clinical studies may provide in understanding dose-effect relationships, linking radiation dose quantities to biological endpoints and even moving away from macro- to microdosimetry. As such, the translation of pre-clinical findings may provide valuable information and new approaches for improved clinical dosimetry, thus paving the way to personalized TAT.
Collapse
Affiliation(s)
- Dayana Castillo Seoane
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium - .,Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium -
| | - Marijke de Saint-Hubert
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Melissa Crabbe
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Lara Struelens
- Research Unit in Dosimetric Applications, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Michel Koole
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), Leuven, Belgium
| |
Collapse
|
243
|
Tran CG, Sherman SK, Howe JR. Small Bowel Neuroendocrine Tumors. Curr Probl Surg 2020; 57:100823. [PMID: 33234227 DOI: 10.1016/j.cpsurg.2020.100823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
| | - Scott K Sherman
- Division of Surgical Oncology and Endocrine Surgery, University of lowa Carver College of Medicine, lowa City, lowa
| | - James R Howe
- Division of Surgical Oncology and Endocrine Surgery, University of lowa Carver College of Medicine, lowa City, lowa.
| |
Collapse
|
244
|
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
|
245
|
Freedman N, Sandström M, Kuten J, Shtraus N, Ospovat I, Schlocker A, Even-Sapir E. Personalized radiation dosimetry for PRRT-how many scans are really required? EJNMMI Phys 2020; 7:26. [PMID: 32394075 PMCID: PMC7214583 DOI: 10.1186/s40658-020-00293-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/27/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose Over recent years, peptide receptor radiotherapy (PRRT) has been recognized as an effective treatment for patients with metastatic neuroendocrine tumors (NETs). Personalized dosimetry can contribute to improve the outcome of peptide receptor radiotherapy (PRRT) in patients with metastatic NETs. Dosimetry can aid treatment planning, ensuring that absorbed dose to vulnerable normal organs (kidneys and bone marrow) does not exceed safe limits over serial treatments, and that absorbed dose to tumor is sufficient. Absorbed dose is estimated from a series of post-treatment SPECT/CT images. Total self-dose is proportional to the integral under the time activity concentration curve (TACC). Method dependence of image-based absorbed dose calculations has been previously investigated, and we set out here to extend previous work by examining implications of number of data points in the TACC and the numerical integration methods used in estimating absorbed dose. Methods In this retrospective study, absorbed dose estimates and effective half-lives were calculated by fitting curves to TACCs for normal organs and tumors in 30 consecutive patients who underwent a series of 4 post-treatment SPECT/CT scans at 4 h, 24 h, 4–5 days, and 1 week following 177Lu-DOTATATE PRRT. We examined the effects of including only 2 or 3 rather than all 4 data points in the TACC, and the effect of numerical integration method (mono-exponential alone or in combination with trapezoidal rule) on the absorbed dose and half-life estimates. Our current method is the combination of trapezoidal rule over the first 24 h, with mono-exponential fit thereafter extrapolated to infinity. The other methods were compared to this current method. Results Differences in absorbed dose and effective half-life between the current method and estimates based only on the second, third, and fourth scans were very small (mean differences < 2.5%), whereas differences between the current method and 4-point mono-exponential fit were higher (mean differences < 5%) with a larger range. It appears that in a 4-point mono-exponential fit the early (4 h) time point may skew results, causing some large errors. Differences between the current method and values based on only 2 time points were relatively small (mean differences < 3.5%) when the 24 h and 1 week scans were used, but when the 24 h and 4–5 days scans, or the 4–5 days and 1 week scans were used, differences were greater. Conclusion This study indicates that for 177Lu-DOTATATE PRRT, accurate estimates of absorbed dose for organs and tumors may be estimated from scans at 24 h, 72 h, and 1 week post-treatment without an earlier scan. It may even be possible to cut out the 72 h scan, though the uncertainty increases. However, further work on more patients is required to validate this.
Collapse
Affiliation(s)
- Nanette Freedman
- Institute of Nuclear Medicine, Tel Aviv Sourasky Medical Center, 6 Weizman Street, 64239, Tel Aviv, Israel.
| | - Mattias Sandström
- Section of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonathan Kuten
- Institute of Nuclear Medicine, Tel Aviv Sourasky Medical Center, 6 Weizman Street, 64239, Tel Aviv, Israel
| | - Natan Shtraus
- Institute of Radiotherapy, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Inna Ospovat
- Institute of Radiotherapy, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Albert Schlocker
- Institute of Radiotherapy, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Einat Even-Sapir
- Institute of Nuclear Medicine, Tel Aviv Sourasky Medical Center, 6 Weizman Street, 64239, Tel Aviv, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
246
|
The five “W”s and “How” of Targeted Alpha Therapy: Why? Who? What? Where? When? and How? RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2020. [DOI: 10.1007/s12210-020-00900-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
247
|
Chun SY, Nguyen MP, Phan TQ, Kim H, Fessler JA, Dewaraja YK. Algorithms and Analyses for Joint Spectral Image Reconstruction in Y-90 Bremsstrahlung SPECT. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:1369-1379. [PMID: 31647425 PMCID: PMC7263381 DOI: 10.1109/tmi.2019.2949068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Quantitative yttrium-90 (Y-90) SPECT imaging is challenging due to the nature of Y-90, an almost pure beta emitter that is associated with a continuous spectrum of bremsstrahlung photons that have a relatively low yield. This paper proposes joint spectral reconstruction (JSR), a novel bremsstrahlung SPECT reconstruction method that uses multiple narrow acquisition windows with accurate multi-band forward modeling to cover a wide range of the energy spectrum. Theoretical analyses using Fisher information and Monte-Carlo (MC) simulation with a digital phantom show that the proposed JSR model with multiple acquisition windows has better performance in terms of covariance (precision) than previous methods using multi-band forward modeling with a single acquisition window, or using a single-band forward modeling with a single acquisition window. We also propose an energy-window subset (ES) algorithm for JSR to achieve fast empirical convergence and maximum-likelihood based initialization for all reconstruction methods to improve quantification accuracy in early iterations. For both MC simulation with a digital phantom and experimental study with a physical multi-sphere phantom, our proposed JSR-ES, a fast algorithm for JSR with ES, yielded higher recovery coefficients (RCs) on hot spheres over all iterations and sphere sizes than all the other evaluated methods, due to fast empirical convergence. In experimental study, for the smallest hot sphere (diameter 1.6cm), at the 20th iteration the increase in RCs with JSR-ES was 66 and 31% compared with single wide and narrow band forward models, respectively. JSR-ES also yielded lower residual count error (RCE) on a cold sphere over all iterations than other methods for MC simulation with known scatter, but led to greater RCE compared with single narrow band forward model at higher iterations for experimental study when using estimated scatter.
Collapse
|
248
|
Lin E, Chen T, Little A, Holliday L, Roach P, Butler P, Hosking E, Bailey E, Elison B, Currow D. Safety and outcomes of 177 Lu-DOTATATE for neuroendocrine tumours: experience in New South Wales, Australia. Intern Med J 2020; 49:1268-1277. [PMID: 31062490 DOI: 10.1111/imj.14336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/14/2019] [Accepted: 04/14/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Peptide receptor radionuclide therapy with 177 Lu-DOTATATE is a promising treatment for inoperable or metastatic neuroendocrine tumours (NET). In 2015, the NSW Ministry of Health provided funding for 177 Lu-DOTATATE treatment of NET under an evaluation framework. AIMS To examine the safety and outcomes of NET patients treated with 177 Lu-DOTATATE under the evaluation framework and assess the statewide implementation of the NSW Lutate therapy referral and protocol for neuroendocrine cancer patients. METHODS A quality of care clinical audit was conducted on all NET patients treated with 177 Lu-DOTATATE from October 2010 to October 2015 at St George Hospital, and from August 2013 to March 2017 at Royal North Shore Hospital. Percentage of patients who met protocol selection criteria was calculated. Survival was estimated using the Kaplan-Meier method. Adjusted regression analyses assessed associations between key clinical factors and outcomes. RESULTS A total of 279 patients was treated. Statewide protocol implementation led to an increase from 60.5 to 83.8% in patients meeting selection criteria. Estimated median overall survival was significantly longer for patients who met selection criteria compared with those who did not (50.7 vs 34.2 months) (P = 0.018). This was driven by the significantly worse overall survival in patients who failed exclusion criteria (P < 0.001). 177 Lu-DOTATATE was well tolerated with haematological, renal and hepatic treatment-related serious adverse events experienced by 9.7, 0.4 and 0.4% of patients respectively. CONCLUSIONS 177 Lu-DOTATATE is a promising treatment for advanced NET. Superior survival in patients who met selection criteria emphasise the importance of protocol adherence.
Collapse
Affiliation(s)
- Enmoore Lin
- Cancer Institute NSW, Sydney, New South Wales, Australia
| | - Tina Chen
- Cancer Institute NSW, Sydney, New South Wales, Australia
| | - Alana Little
- Cancer Institute NSW, Sydney, New South Wales, Australia
| | - Laura Holliday
- Cancer Institute NSW, Sydney, New South Wales, Australia
| | - Paul Roach
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Patrick Butler
- Department of Nuclear Medicine, St George Hospital, Sydney, New South Wales, Australia
| | - Erika Hosking
- Department of Nuclear Medicine, St George Hospital, Sydney, New South Wales, Australia
| | - Elizabeth Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Barry Elison
- Agency for Clinical Innovation, Sydney, New South Wales, Australia
| | - David Currow
- Cancer Institute NSW, Sydney, New South Wales, Australia
| |
Collapse
|
249
|
Ramonaheng K, van Staden JA, du Raan H. Validation of a Monte Carlo modelled gamma camera for Lutetium-177 imaging. Appl Radiat Isot 2020; 163:109200. [PMID: 32561041 DOI: 10.1016/j.apradiso.2020.109200] [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] [Received: 10/15/2019] [Revised: 02/11/2020] [Accepted: 04/08/2020] [Indexed: 10/24/2022]
Abstract
This study validated a model of the Siemens Symbia T16 dual-head SPECT/CT gamma camera created using the Monte Carlo program SIMIND for 177Lu. The validation was done by comparing experimental and simulated gamma camera performance criteria tests for the 177Lu 208 keV photopeak with a medium-energy collimator. Results showed good agreement between the experimental and simulated values. These results illustrated that SIMIND could emulate the Symbia T16 successfully and therefore, can be used with confidence to model 177Lu images.
Collapse
Affiliation(s)
- K Ramonaheng
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa.
| | - J A van Staden
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| | - H du Raan
- Department of Medical Physics, University of the Free State, PO Box 339, Bloemfontein, 9300, South Africa
| |
Collapse
|
250
|
Abou Jokh Casas E, Pubul Núñez V, Anido-Herranz U, del Carmen Mallón Araujo M, del Carmen Pombo Pasín M, Garrido Pumar M, Cabezas Agrícola JM, Cameselle-Teijeiro JM, Hilal A, Ruibal Morell Á. Evaluation of 177Lu-Dotatate treatment in patients with metastatic neuroendocrine tumors and prognostic factors. World J Gastroenterol 2020; 26:1513-1524. [PMID: 32308351 PMCID: PMC7152518 DOI: 10.3748/wjg.v26.i13.1513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/06/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND 177Lu peptide receptor radionuclide therapy (PRRT) is a recently approved therapy in Spain that has been demonstrated to be a well-tolerated therapy for positive somatostatin receptor advanced gastroenteropancreatic neuroendocrine tumors.
AIM To determine the impact of PRRT on quality of life, radiologic and metabolic response, overall survival, prognostic factors and toxicity.
METHODS Thirty-six patients treated with 177Lu-PRRT from 2016 to 2019 were included. The most frequent location of the primary tumor was the gastrointestinal tract (52.8%), pancreas (27.8%), and nongastropancreatic neuroendocrine tumor (11.1%). The liver was the most common site of metastasis (91.7%), followed by distant nodes (50.0%), bone (27.8%), peritoneum (25.0%) and lung (11.1%). Toxicity was evaluated after the administration of each dose. Treatment efficacy was evaluated by two parameters: stable disease and disease progression in response evaluation criteria in solid tumors 1.1 criterion and prognostic factors were tested.
RESULTS From 36 patients, 55.6% were men, with a median age of 61.1 ± 11.8 years. Regarding previous treatments, 55.6% of patients underwent surgery of the primary tumor, 100% of patients were treated with long-acting somatostatin analogues, 66.7% of patients were treated with everolimus, 27.8% of patients were treated with tyrosine kinase inhibitor, and 27.8% of patients were treated with interferon. One patient received radioembolization, three patients received chemoembolization, six patients received chemotherapy. Hematological toxicity was registered in 14 patients (G1-G2: 55.5% and G3: 3.1%). Other events presented were intestinal suboclusion in 4 cases, cholestasis in 2 cases and carcinoid crisis in 1 case. The median follow-up time was 3 years. Currently, 24 patients completed treatment. Nineteen are alive with stable disease, two have disease progression, eight have died, and nine are still receiving treatment. The median overall survival was 12.5 mo (95% confidence interval range: 9.8–15.2), being inversely proportional to toxicity in previous treatments (P < 0.02), tumor grade (P < 0.01) and the presence of bone lesions (P = 0.009) and directly proportional with matching lesion findings between Octreoscan and computed tomography pre-PRRT (P < 0.01), , primary tumor surgery (P = 0.03) and metastasis surgery (P = 0.045). In a multivariate Cox regression analysis, a high Ki67 index (P = 0.003), a mismatch in the lesion findings between Octreoscan and computed tomography pre-PRRT (P < 0.01) and a preceding toxicity in previous treatments (P < 0.05) were risk factors to overall survival.
CONCLUSION Overall survival was inversely proportional to previous toxicity, tumor grade and the presence of bone metastasis and directly proportional to matching lesion findings between Octreoscan and computed tomography pre-PRRT and primary tumor and metastasis surgery.
Collapse
Affiliation(s)
- Estephany Abou Jokh Casas
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Virginia Pubul Núñez
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Urbano Anido-Herranz
- Department of Oncology, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - María del Carmen Mallón Araujo
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Maria del Carmen Pombo Pasín
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Miguel Garrido Pumar
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - José Manuel Cabezas Agrícola
- Department of Endocrinology, Santiago de Compostela´s University Hospital, Santiago de Compostela, 15706, A Coruña, Spain
| | | | - Ashraf Hilal
- Department of Statistics, University of Santiago de Compostela, Santiago de Compostela 15706, A Coruña, Spain
| | - Álvaro Ruibal Morell
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| |
Collapse
|