1
|
Quinn M, Kemkem Y, White G, Touska P, Christodoulou D, Jacques A, Breen L, McGowan B, Joshi M, Ul Hassan F, Harrison-Phipps K, Hubbard JG, Obholzer R, Izatt L, Carroll P, Velusamy A. Clinical, genetic, radiological characteristics and management of mediastinal paragangliomas: a literature review and case series. Endocr Relat Cancer 2025; 32:e240279. [PMID: 40063004 PMCID: PMC11964478 DOI: 10.1530/erc-24-0279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
Paragangliomas (PGLs) are neuroendocrine tumours (NETs) that arise from neural crest-derived cells. Up to 40% of cases occur due to the presence of a pathogenic germline variant (PGV) in a known gene. Mediastinal PGLs are rare but are being diagnosed with increasing frequency. Treatment generally involves surgery but is complicated in mediastinal PGLs due to their anatomy. Here, we will perform a literature review and discuss our experience with 18 such cases. Cases were identified via the Guy's and St Thomas' NHS Foundation Trust NET multidisciplinary team database. Tumours ranged in size from 0.6 × 0.6 to 6.8 × 4.9 cm. 72.2% were associated with a PGV of SDHB or SDHD. 22.2% developed metastatic disease, but it was only possible to attribute 50% of these to a mediastinal primary. 68Ga-DOTATATE PET CT demonstrated 100% sensitivity. The literature review identified 233 cases. A PGV was reported in 81% of cases, with metastatic disease in approximately 39.2%. It was not possible to confirm that all cases of metastatic disease were secondary to a mediastinal primary. Our experience confirms the high rate of mediastinal PGLs arising in the presence of a PGV. The lower rate of metastatic disease in our cohort (11.1%) likely represents earlier diagnosis thanks to the application of screening protocols and the increased sensitivity of 68Ga-DOTATATE PET CT. With this increased sensitivity, we have diagnosed small mediastinal PGLs that were not evident on alternative imaging modalities. In the absence of growth or catecholamine secretion, the need to intervene on these is unclear.
Collapse
Affiliation(s)
- Mark Quinn
- Departments of Endocrinology, Radiology, Nuclear Medicine, Cardio-Thoracic Surgery, Genetics, Endocrine Surgery and ENT, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Santo G, di Santo G, Cicone F, Virgolini I. Peptide receptor radionuclide therapy with somatostatin analogs beyond gastroenteropancreatic neuroendocrine tumors. J Neuroendocrinol 2025; 37:e70013. [PMID: 40064181 PMCID: PMC11919479 DOI: 10.1111/jne.70013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
First isolated by Brazeau et al. in 1972, somatostatin (SST) is a neuropeptide known for regulating various signaling pathways through its specific cell surface receptors. Somatostatin receptors (SSTRs) comprise a family of five G protein-coupled receptors that are widely distributed across the human body and are expressed by various tumor types. The growing understanding of their clinical potential led to the introduction of both cold and radiolabeled somatostatin analogs (SSAs), which have revolutionized the management of several cancers, especially neuroendocrine tumors. As a direct consequence, advances in peptide receptor radionuclide therapy (PRRT) over the last 30 years led to the approval of 177Lu-DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors (GEPNETs). Theoretically, any cancer patients whose tumors express SSTR, as demonstrated in vivo through SSTR-based molecular imaging, could be candidates for PRRT, especially those with limited treatment options. However, evidence on the efficacy of PRRT in non-GEPNET SSTR-expressing tumors is limited, and mainly derived from small retrospective studies. Given the limited therapeutic options for advanced/metastatic patients, there is a clear need for randomized trials to formally approve PRRT with SSAs for patients who may benefit from this treatment, particularly in certain types of neuroendocrine neoplasms such as lung carcinoids, paragangliomas, and meningiomas, where high rates of disease control (up to 80%) can be achieved. In addition, emerging evidence supports the potential of combination therapies, alpha emitters, and non-SSTR-based radionuclide therapy in tumors beyond GEPNET. This review aims to provide a comprehensive overview of PRRT's role in cancers beyond GEPNET, exploring new possibilities and future directions for most SSTR highly expressing tumors.
Collapse
Affiliation(s)
- Giulia Santo
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
- Department of Experimental and Clinical Medicine“Magna Graecia” University of CatanzaroCatanzaroItaly
| | - Gianpaolo di Santo
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine“Magna Graecia” University of CatanzaroCatanzaroItaly
| | - Irene Virgolini
- Department of Nuclear MedicineMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
3
|
Badrane I, Urso L, Campennì A, Cittanti C, De Rimini ML, Bartolomei M. Radioligand therapy in sympathetic-adrenal-medullary axis tumors: state of art and perspectives. Endocrine 2025; 87:67-72. [PMID: 39373830 DOI: 10.1007/s12020-024-04062-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024]
Abstract
The approval in 2017 by the European Medicines Agency (EMA) and in 2018 by the US Food and Drug Administration (FDA) of radioligand therapy (RLT) led to its wide application in therapeutic management of neuroendocrine neoplasms (NENs). However, the indications are currently limited to certain specific histotypes belonging to the broader NEN's family, mainly advanced well-differentiated gastro-entero-pancreatic NENs. As a consequence, several other tumors of the NEN spectrum that can potentially benefit, due to their biological characteristics, from RLT are still ineligible and can be considered "RLT-orphans". Among those, the subgroup of NENs originating from the sympathetic-adrenal-medullary (SAM) axis can be listed. This paper discusses the state of art and perspectives of the theragnostic applications in pheochromocytomas and paragangliomas, considering both the traditional theragnostic model - with radiolabelled metaiodobenzylguanidine (MIBG) - and the innovative one with radiolabeled somatostatin analogs (SSAs), that will hopefully become available for these patients in the near future.
Collapse
Affiliation(s)
- Ilham Badrane
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Nuclear Medicine Unit, Onco-Hematology Department, University Hospital of Ferrara, Ferarra, Italy
| | - Luca Urso
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Nuclear Medicine Unit, Onco-Hematology Department, University Hospital of Ferrara, Ferarra, Italy
| | - Alfredo Campennì
- Unit of Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy.
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Nuclear Medicine Unit, Onco-Hematology Department, University Hospital of Ferrara, Ferarra, Italy
| | - Maria Luisa De Rimini
- Unit of Nuclear Medicine, Department of Health Service, AO Colli Hospitals, Naples, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Onco-Hematology Department, University Hospital of Ferrara, Ferarra, Italy
| |
Collapse
|
4
|
Gild ML, Do K, Tsang VHM, Tacon LJ, Clifton-Bligh RJ, Robinson BG. Pheochromocytoma in MEN2. Recent Results Cancer Res 2025; 223:211-235. [PMID: 40102259 DOI: 10.1007/978-3-031-80396-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Pheochromocytomas (PCs) are rare neuroendocrine tumors found in 20-50% of MEN2 patients. MEN2-related PCs are more often bilateral, identified at a younger age and have a low metastatic potential. They secrete epinephrine as the predominant catecholamine, along with its metabolite metanephrine, and lesser amounts of norepinephrine and normetanephrine. The advent of molecular diagnostic tools has enhanced the identification and stratification of these tumors, revealing a strong genotype-phenotype correlation which is crucial for screening and managing patients. Evaluation involves a combination of structural (CT/MRI) and functional imaging. MIBG remains helpful for PC assessment but novel PET ligands (18F-DOPA, 68Ga-DOTATATE, 18F-FDG) aid in the detection of extra-adrenal paragangliomas, recurrence, and metastatic disease. The treatment paradigm has shifted toward personalized medicine, incorporating genetic insights to tailor interventions, particularly surgical approaches and novel therapeutics such as radiolabeling of somatostatin analogs with lutetium and tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Matti L Gild
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia.
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia.
| | - Kimchi Do
- Department of Endocrinology, Nepean Hospital, Sydney, Australia
| | - Venessa H M Tsang
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Lyndal J Tacon
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Roderick J Clifton-Bligh
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Bruce G Robinson
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| |
Collapse
|
5
|
Varghese J, Skefos CM, Jimenez C. Metastatic pheochromocytoma and paraganglioma: Integrating tumor biology in clinical practice. Mol Cell Endocrinol 2024; 592:112344. [PMID: 39182716 DOI: 10.1016/j.mce.2024.112344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumors derived from chromaffin cells in the autonomic nervous system. Depending on their location, these tumors are capable of excessive catecholamine production, which may lead to uncontrolled hypertension and other life-threatening complications. They are associated with a significant risk of metastatic disease and are often caused by an inherited germline mutation. Although surgery can cure localized disease and lead to remission, treatments for metastatic PPGL (mPPGL)-including chemotherapy, radiopharmaceutical agents, multikinase inhibitors, and immunotherapy used alone or in combination- aim to control tumor growth and limit organ damage. Substantial advances have been made in understanding hereditary and somatic molecular signaling pathways that play a role in tumor growth and metastasis. Treatment options for metastatic disease are rapidly evolving, and this paper aims to provide a brief overview of the management of mPPGL with a focus on therapy options.
Collapse
Affiliation(s)
- Jeena Varghese
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Catherine M Skefos
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
6
|
Severi S, Grassi I, Bongiovanni A, Nicolini S, Marini I, Arpa D, Ranallo N, Azzali I, Di Iorio V, Sarnelli A, Manuela M, Amadori E, Fabbri L, Bartolini D, Tosatto L, Di Meco F, Gurrieri L, Riva N, Calabro L, Matteucci F, Paganelli G, Sansovini M. Peptide Receptor Radionuclide Therapy in Advanced Refractory Meningiomas: Efficacy and Toxicity in a Long Follow-up. J Nucl Med 2024; 65:1409-1415. [PMID: 39142827 PMCID: PMC11372258 DOI: 10.2967/jnumed.123.266956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/25/2024] [Indexed: 08/16/2024] Open
Abstract
Recurrence of meningiomas after surgery and radiotherapy deserves specific attention because of the lack of active third-line therapies. Somatostatin receptors are usually overexpressed on the cell membrane of meningiomas, and this has led the way to a radionuclide theranostic approach. Diagnoses with 68Ga-DOTA-octreotide and peptide receptor radionuclide therapy (PRRT) with 90Y/177Lu-DOTA-octreotide are currently possible options within experimental protocols or as compassionate use in small patient groups. Methods: From October 2009 to October 2021, 42 meningioma patients with radiologic recurrence after standard therapies were treated with 90Y-DOTATOC (dosage of 1.1 or 5.5 GBq) or with 177Lu-DOTATATE (dosage of 3.7 or 5.5 GBq) in a mean of 4 cycles. All patients showed intense uptake at diagnostic 68Ga-DOTATOC PET/CT or in an 111In-octreotide scan. Results: Of 42 patients treated, 5 patients received 90Y-DOTATOC with a cumulative activity of 11.1 GBq and 37 patients received 177Lu-DOTATATE with a cumulative activity of 22 GBq. The disease control rate was 57%. With a median follow-up of 63 mo, median progression-free survival was 16 mo, and median overall survival was 36 mo. Retreatment 177Lu-PRRT was performed in 6 patients with an administered median activity of 13 GBq in a mean of 5 cycles. With a 75.8-mo follow-up, median progression-free survival and overall survival were 6.5 and 17 mo, respectively. Only 1 patient discontinued the treatment because of grade 3 platelet toxicity. A rapidly transient grade 2 neutropenia was recorded in 1 retreated patient. Conclusion: PRRT in patients with advanced meningiomas overexpressing somatostatin receptor 2 was active and well tolerated, showing a 57% disease control rate. Furthermore, PRRT could represent a potential retreatment option. Further studies, also in combination with other treatments, are warranted.
Collapse
Affiliation(s)
- Stefano Severi
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Ilaria Grassi
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumor Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy;
| | - Silvia Nicolini
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Irene Marini
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Donatella Arpa
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Emilia Romagna, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumor Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Irene Azzali
- Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Valentina Di Iorio
- Oncological Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Emilia Romagna, Italy
| | - Anna Sarnelli
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Monti Manuela
- Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Elena Amadori
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Lucia Fabbri
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Emilia Romagna, Italy
| | | | - Luigino Tosatto
- Department of Neurosciences, Neurosurgery Division "M Bufalini" Hospital, Cesena, Emilia Romagna, Italy
| | - Francesco Di Meco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Lorena Gurrieri
- Osteoncology and Rare Tumor Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Nada Riva
- Osteoncology and Rare Tumor Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Luana Calabro
- Department of Oncology, University Hospital of Ferrara, Cona, Italy; and
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Federica Matteucci
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| | - Maddalena Sansovini
- Nuclear Medicine and Radiometabolic Units, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori", Meldola, Italy
| |
Collapse
|
7
|
Nasca V, Prinzi N, Coppa J, Prisciandaro M, Oldani S, Ghelardi F, Conca E, Capone I, Busico A, Perrone F, Tamborini E, Sabella G, Greco G, Greco FG, Tafuto S, Procopio G, Morano F, Niger M, Maccauro M, Milione M, de Braud F, Pietrantonio F, Pusceddu S. Sunitinib for the treatment of patients with advanced pheochromocytomas or paragangliomas: The phase 2 non-randomized SUTNET clinical trial. Eur J Cancer 2024; 209:114276. [PMID: 39128186 DOI: 10.1016/j.ejca.2024.114276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Metastatic Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors characterized by high morbidity and limited systemic treatment options, mainly based on radiometabolic treatments or chemotherapy. Based on the preclinical rationale that PGGLs carcinogenesis relies on angiogenesis, treatment with tyrosine kinase inhibitors (TKI) may represent another viable therapeutic option. METHODS We conducted a prospective phase II study in patients with metastatic or unresectable PGGLs. Patients received sunitinib (50 mg daily for 4 weeks, followed by a 2-week rest period) until progressive disease (PD), unacceptable toxicity or consent withdrawal. The primary endpoint was 12-month progression-free survival (PFS) rate; secondary endpoints were safety overall response rate (ORR) according to RECIST 1.1 criteria and overall survival (OS). EudraCT Number: 2011-002632-99. RESULTS Fifty patients were included. At a median follow-up of 71.7 months (IQR 35.4-100.1), the 1 year-PFS rate was 53.4 % (95 %CI 41.1-69.3) and median PFS was 14.1 months (95 % CI 8.9-25.7). ORR was 15.6 %, the median OS was 49.4 months (95 %CI 21.2-NA), and grade 3 or higher treatment-related adverse events were reported in 34 % patients. No significant correlation was found between specific genetic alterations or genomic clusters and sunitinib efficacy. CONCLUSION Sunitinib is an active drug in patients with advanced PGGLs, capable of inducing prolonged disease control with a manageable toxicity profile.
Collapse
Affiliation(s)
- Vincenzo Nasca
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Natalie Prinzi
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Jorgelina Coppa
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Prisciandaro
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Simone Oldani
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Filippo Ghelardi
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Elena Conca
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Iolanda Capone
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Adele Busico
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Tamborini
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanna Sabella
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgio Greco
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Salvatore Tafuto
- Sarcoma and Rare Tumors Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy; ENETs Center of Excellence, Naples, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Marco Maccauro
- Department of Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, ENETS Center of Excellence, Milan, Italy
| | - Massimo Milione
- Diagnostic Pathology and Laboratory Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy; University of Milan, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy; ENETS Center of Excellence, Fondazione IRCCS Istituto Tumori Milano, Milan, Italy.
| |
Collapse
|
8
|
Sukrithan V, Perez K, Pandit-Taskar N, Jimenez C. Management of metastatic pheochromocytomas and paragangliomas: when and what. Curr Probl Cancer 2024; 51:101116. [PMID: 39024846 DOI: 10.1016/j.currproblcancer.2024.101116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Recently, the treatment landscape for metastatic pheochromocytomas and paragangliomas (MPPGL) has seen both progress and setbacks. We provide an up-to-date review of the multimodality management of MPPGL and discuss novel opportunities and current challenges in the treatment landscape. Given the unique clinical presentation of MPPGL, we discuss the management of hormone-related clinical sequelae and traditional modalities of therapy. Advances in the understanding of the molecular biology of these diverse tumors have enabled novel strategies such as augmenting DNA damage by targeted delivery of radionuclides such as 131I and 177Lu, abrogating tumor angiogenesis, hypoxia resistance, and DNA damage repair. Despite progress, we address the significant challenges still faced by patients and researchers engaged in efforts to improve outcomes in these rare cancers.
Collapse
Affiliation(s)
- Vineeth Sukrithan
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Neeta Pandit-Taskar
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Shen Y, Luo Y, Li M, Luo R, Chen L, Gao X, Jiang J, Liu Y, Lu Z, Zhang J. Somatostatin receptor subtype 2A expression and genetics in 184 paragangliomas: a single center retrospective observational study. Endocrine 2024; 85:398-406. [PMID: 38306009 DOI: 10.1007/s12020-023-03595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/01/2023] [Indexed: 02/03/2024]
Abstract
PURPOSE Adrenal and extra-adrenal paragangliomas (PGLs) are a group of neuroendocrine tumors (NETs) with strong heterogeneity, which often express somatostatin receptor subtype 2 A (SSTR2A). However, the association between SSTR2A expression and genetic status of PGLs remains unclear. The purpose of the study was to identify whether various pathogenic variants (PVs) had an impact on SSTR2A expression in PGLs. METHODS This retrospective study included 184 patients with pathologically confirmed PGLs. The immunohistochemical expression of SSTR2A were studied in 184 tumors and PVs were tested in 159 tumor samples. Clinical and genetic data were compared in SSTR2A positive and negative PGLs. RESULTS SSTR2A was positive in 63.6% (117/184) of all tumors. PGLs with negative SSTR2A were more likely to be extra-adrenal (37.0% vs 18.0%; P = 0.005) and exhibited a considerably greater proportion of PVs (75.4% vs. 49.0%; P = 0.001) than those with positive SSTR2A. Compared to those without PVs, a higher proportion of PGLs with PVs in cluster 1B (P = 0.004) and cluster 2 (P = 0.004) genes, especially VHL (P = 0.009), FGFR1 (P = 0.010) and HRAS (P = 0.007), were SSTR2A negative. SSTR2A was positive in all tumors (4/4) with SDHx PVs and in 87.5% (7/8) of metastatic PGLs. CONCLUSIONS SSTR2A negativity was correlated with extra-adrenal tumor location and PVs in cluster 1B and cluster 2 genes such as VHL, FGFR1 and HRAS. Immunohistochemistry of SSTR2A should be taken into consideration in the personalized management of PGLs.
Collapse
Affiliation(s)
- Yanting Shen
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Yu Luo
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minghao Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lingli Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Yujun Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Zhiqiang Lu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Jing Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China.
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Yang H, Zhang Y, Li H, Zhang Y, Feng Y, Yang X, Chen Y. Efficacy and Safety of 225 Ac-DOTATATE in the Treatment of Neuroendocrine Neoplasms With High SSTR Expression. Clin Nucl Med 2024; 49:505-512. [PMID: 38498615 DOI: 10.1097/rlu.0000000000005149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
PURPOSE We aimed to evaluate the efficacy and safety of 225 Ac-DOTATATE targeted α therapy (TAT) in various neuroendocrine neoplasms (NENs) with high somatostatin receptor (SSTR) expression. PATIENTS AND METHODS This single-center prospective study included 10 patients with histologically diagnosed NENs that exhibited increased SSTR expression on 68 Ga-DOTATATE PET/CT imaging. All patients received 225 Ac-DOTATATE TAT. The primary end points were molecular imaging-based response and disease control rate (DCR), measured using the slightly modified Positron Emission Tomography Response Criteria in Solid Tumors 1.0. The secondary end points were adverse event profiles and clinical responses. The adverse event profile was determined according to the Common Terminology Criteria for Adverse Events version 5.0. Clinical response was assessed using the EORTC QLQ-C30 v3.0 (European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire version 3.0). RESULTS A molecular imaging-based partial response was observed in 40% of all patients, SD in 40%, PD in 20%, and DCR in 80%. The DCR was 83.3% (5/6) in patients who were previously treated with 177 Lu-DOTATATE. According to the EORTC QLQ-C30 v3.0 score, most symptoms improved after 225 Ac-DOTATATE treatment, with only diarrhea showing no improvement. Grade III/IV hematological, kidney, and liver toxicities were not observed. The median follow-up time was 14 months (7-22 months), and no deaths were reported. CONCLUSIONS This initial study suggests that 225 Ac-DOTATATE is a potentially promising option for treating NENs with elevated SSTR expression, with an acceptable toxicity profile and well-tolerated adverse effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiqun Yang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | |
Collapse
|
11
|
Palade DO, Hainarosie R, Zamfir A, Vrinceanu D, Pertea M, Tusaliu M, Mocanu F, Voiosu C. Paragangliomas of the Head and Neck: A Review of the Latest Diagnostic and Treatment Methods. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:914. [PMID: 38929531 PMCID: PMC11205799 DOI: 10.3390/medicina60060914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Background and objectives: Paragangliomas of the head and neck are rare, slow-growing neuroendocrine tumors, benign in their vast majority, but with a possibility of developing distant metastases. They show great inheritable character, and their behavior has proven to be unpredictable; therefore, they are considered malignant. Material and methods: This article aims to offer a more comprehensive presentation of the pathogenesis, epidemiology, diagnostic methods, imaging development, and treatment guidelines. We tried to bring together all the necessary data that, in our opinion, a head and neck practitioner should know when managing this type of tumor. Our main focus is on the most recent studies, with the purpose of a homogenous presentation of all current guidelines and approaches to this pathology. Results: Paragangliomas of the head and neck are still a disputed topic. One of the main reasons for that is their low incidence of 0.3 to 1 per 100,000 every year. The most frequent locations are the carotid body, the temporal bone, the jugular and mastoid foramen, and the vagal nerve. Their clinical presentation usually involves a painless lateral mass associated with symptoms such as hoarseness, hearing loss, tinnitus, and cranial nerve deficits. Up to 40% of them are inherited, mostly linked with mutations of succinate dehydrogenase complex. Imaging evaluation consists of CT and MRI, and new functional explorations such as 18F-FDA and 18F-FDG PET/CT, 18F-DOPA PET, 123I-MIBG, and 68Ga-DOTATE PET/CT. Measuring the catecholamine levels in the plasma and urine is mandatory, even though paragangliomas of the head and neck rarely display secretory behavior. Treatment mainly consists of surgery, with different approaches and techniques, but conservative management methods such as wait and scan, radiotherapy, proton therapy, and chemotherapy have proven their efficiency. The therapeutical decision lacks consensus, and current studies tend to recommend an individualized approach. Guidelines regarding long-term follow-up are still a matter of debate.
Collapse
Affiliation(s)
- Dragos Octavian Palade
- Surgery Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- ENT Department, “Sf. Spiridon” Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Razvan Hainarosie
- ENT Department, Faculty of Medicine, “Carol Davilla” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Adina Zamfir
- ENT Department, “Grigore Alexandrescu” Children’s Emergency Hospital, 011743 Bucharest, Romania
| | - Daniela Vrinceanu
- ENT Department, Faculty of Medicine, “Carol Davilla” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihaela Pertea
- Surgery Department, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihail Tusaliu
- ENT Department, Faculty of Medicine, “Carol Davilla” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Florin Mocanu
- ENT Department, “Sf. Spiridon” Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Catalina Voiosu
- ENT Department, Faculty of Medicine, “Carol Davilla” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
12
|
Timmers HJLM, Taïeb D, Pacak K, Lenders JWM. Imaging of Pheochromocytomas and Paragangliomas. Endocr Rev 2024; 45:414-434. [PMID: 38206185 PMCID: PMC11074798 DOI: 10.1210/endrev/bnae001] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Pheochromocytomas/paragangliomas are unique in their highly variable molecular landscape driven by genetic alterations, either germline or somatic. These mutations translate into different clusters with distinct tumor locations, biochemical/metabolomic features, tumor cell characteristics (eg, receptors, transporters), and disease course. Such tumor heterogeneity calls for different imaging strategies in order to provide proper diagnosis and follow-up. This also warrants selection of the most appropriate and locally available imaging modalities tailored to an individual patient based on consideration of many relevant factors including age, (anticipated) tumor location(s), size, and multifocality, underlying genotype, biochemical phenotype, chance of metastases, as well as the patient's personal preference and treatment goals. Anatomical imaging using computed tomography and magnetic resonance imaging and functional imaging using positron emission tomography and single photon emission computed tomography are currently a cornerstone in the evaluation of patients with pheochromocytomas/paragangliomas. In modern nuclear medicine practice, a multitude of radionuclides with relevance to diagnostic work-up and treatment planning (theranostics) is available, including radiolabeled metaiodobenzylguanidine, fluorodeoxyglucose, fluorodihydroxyphenylalanine, and somatostatin analogues. This review amalgamates up-to-date imaging guidelines, expert opinions, and recent discoveries. Based on the rich toolbox for anatomical and functional imaging that is currently available, we aim to define a customized approach in patients with (suspected) pheochromocytomas/paragangliomas from a practical clinical perspective. We provide imaging algorithms for different starting points for initial diagnostic work-up and course of the disease, including adrenal incidentaloma, established biochemical diagnosis, postsurgical follow-up, tumor screening in pathogenic variant carriers, staging and restaging of metastatic disease, theranostics, and response monitoring.
Collapse
Affiliation(s)
- Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France and European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1583, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
13
|
Rubino M, Di Stasio GD, Bodei L, Papi S, Rocca PA, Ferrari ME, Fodor CI, Bagnardi V, Frassoni S, Mei R, Fazio N, Ceci F, Grana CM. Peptide receptor radionuclide therapy with 177Lu- or 90Y-SSTR peptides in malignant pheochromocytomas (PCCs) and paragangliomas (PGLs): results from a single institutional retrospective analysis. Endocrine 2024; 84:704-710. [PMID: 38324106 PMCID: PMC12054632 DOI: 10.1007/s12020-024-03707-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/18/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Malignant pheochromocytomas (PCCs) and paragangliomas (PGLs) are rare tumors and available systemic therapies are limited. AIM To explore the role of peptide receptor radionuclide therapy (PRRT) with Yttrium-90 (90Y) and Lutetium-177 (177Lu) peptides in pheochromocytomas (PCCs) and paragangliomas (PGLs). METHODS We retrospectively analyzed more than 1500 patients with histologically proven neuroendocrine tumors treated with 177Lu- or 90Y-DOTA-TATE or -TOC between 1999 to 2017 at our Institute. Overall, 30 patients with confirmed malignant PCCs and PGLs matched inclusion/exclusion criteria and were considered eligible for this analysis. RESULTS Thirty (n = 30) patients were treated: 22 with PGLs and 8 with PCCs (12 M and 18 F, median age 47 [IQR: 35-60 years]). Eighteen patients (n = 18) had head and neck PGLs, 3 patients thoracic PGLs and 1 patient abdominal PGL. Sixteen patients (53%) had locally advanced and fourteen (47%) had metastatic disease. Twenty-seven (90%) patients had disease progression at baseline. Four (13%) patients were treated with 90Y, sixteen (53%) with 177Lu and ten (33%) with 90Y + 177Lu respectively. The median total cumulative activity from treatment with 90Y- alone was 9.45 GBq (range 5.11-14.02 GBq), from 177Lu- alone was 21.9 GBq (7.55-32.12 GBq) and from the combination treatment was 4.94 GBq from 90Y- and 6.83 GBq from 177Lu- (ranges 1.04-10.1 and 2.66-20.13 GBq, respectively). Seven out of 30 (23%) patients had partial response and 19 (63%) stable disease. Median follow up was 8.9 years (IQR: 2.9-12). The 5-y and 10-y PFS was 68% (95% CI: 48-82) and 53% (95% CI: 33-69), respectively, whereas 5-y and 10-y OS was 75% (95% CI: 54-87) and 59% (95% CI: 38-75), respectively. Grade 3 or 4 acute hematological toxicity occurred in three patients, two with leucopenia and one with thrombocytopenia, respectively. CONCLUSION PRRT with 177Lu- or 90Y-DOTA-TATE or -TOC is feasible and well tolerated in advanced PGLs and PCCs.
Collapse
Affiliation(s)
- Manila Rubino
- Onco-Endocrinology Unit, IEO European Institute of Oncology IRCCS, Milano, Italy
| | | | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefano Papi
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - Paola Anna Rocca
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milano, Italy
| | | | - Cristiana Iuliana Fodor
- Division of Radiotherapy, Data Management, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milano, Italy
| | - Samuele Frassoni
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milano, Italy
| | - Riccardo Mei
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumours, IEO European Institute of Oncology, IRCCS, Milano, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Italy
| | - Chiara Maria Grana
- Radiometabolic Therapy Unit, Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milano, Italy.
| |
Collapse
|
14
|
Mallak N, O'Brien SR, Pryma DA, Mittra E. Theranostics in Neuroendocrine Tumors. Cancer J 2024; 30:185-193. [PMID: 38753753 DOI: 10.1097/ppo.0000000000000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Neuroendocrine tumors (NETs) are rare tumors that develop from cells of the neuroendocrine system and can originate in multiple organs and tissues such as the bowels, pancreas, adrenal glands, ganglia, thyroid, and lungs. This review will focus on gastroenteropancreatic NETs (more commonly called NETs) characterized by frequent somatostatin receptor (SSTR) overexpression and pheochromocytomas/paragangliomas (PPGLs), which typically overexpress norepinephrine transporter. Advancements in SSTR-targeted imaging and treatment have revolutionized the management of patients with NETs. This comprehensive review delves into the current practice, discussing the use of the various Food and Drug Administration-approved SSTR-agonist positron emission tomography tracers and the predictive imaging biomarkers, and elaborating on 177Lu-DOTATATE peptide receptor radionuclide therapy including the evolving areas of posttherapy imaging practices and peptide receptor radionuclide therapy retreatment. SSTR-targeted imaging and therapy can also be used in patients with PPGL; however, this patient population has demonstrated the best outcomes from norepinephrine transporter-targeted therapy with 131I-metaiodobenzylguanidine. Metaiodobenzylguanidine theranostics for PPGL will be discussed, noting that in 2024 it became commercially unavailable in the United States. Therefore, the use and reported success of SSTR theranostics for PPGL will also be explored.
Collapse
Affiliation(s)
- Nadine Mallak
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| | - Sophia R O'Brien
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Daniel A Pryma
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Erik Mittra
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| |
Collapse
|
15
|
Tian R, Yao X, Song J, Wang J, Fu J, Shi L, Yu F, Zhang P, Zhang C, Ni Y, Wang F. Anlotinib for Metastatic Progressed Pheochromocytoma and Paraganglioma: A Retrospective Study of Real-World Data. J Endocr Soc 2024; 8:bvae061. [PMID: 38650712 PMCID: PMC11033215 DOI: 10.1210/jendso/bvae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Indexed: 04/25/2024] Open
Abstract
Introduction Pheochromocytomas (PCC) and paragangliomas (PGL) (collectively PPGL) are a type of rare hypervascular neuroendocrine tumors that are very challenging to treat. This study aimed to determine the efficacy and safety of the multi-tyrosine kinase inhibitor anlotinib for the treatment of locally advanced or metastatic (LA/M) PPGL. Methods A total of 37 eligible patients with unresectable or progressive LA/M PPGL were enrolled. Of them, 27 patients received anlotinib alone (n = 19) or in combination (n = 8) with radionuclide therapies, including peptide receptor radionuclide therapy (PRRT) and iodine 131 meta-iodobenzylguanidine (131I-MIBG). The primary endpoints included objective response rate (ORR), defined as partial response (PR) or complete response (CR), and disease-control rate, defined as PR, CR, or stable disease (SD). The secondary endpoints were progression-free survival (PFS), duration of response, and drug safety. Results In the efficacy evaluation for all 27 patients, the ORR was 44.44% (95% CI: 24.4%-64.5%) and disease-control rate was 96.29% (95% CI: 88.7%-100%). Twelve cases (44.44%) achieved PR, 14 (51.85%) SD. The median PFS was 25.2 months (95% CI: 17.2 months to not reached). PFS was shorter in the anlotinib monotherapy group than in the group receiving anlotinib in combination with radionuclide therapy (P = .2). There were no serious treatment-related AEs. Conclusion Anlotinib monotherapy or in combination with radionuclide therapies shows promising efficacy and safety for the treatment of LA/M PCC and PGL. Multi-tyrosine kinase inhibitors might represent a novel therapeutic strategy for patients with PPGL; however, large-scale prospective randomized, blinded, controlled clinical research studies are required.
Collapse
Affiliation(s)
- Rui Tian
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaochen Yao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jieping Song
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jun Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingjing Fu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Liang Shi
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Fei Yu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Pengjun Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chuan Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yudan Ni
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
16
|
Su D, Zhang Y, Li Z, Yang H, Chen Y. 177 Lu-DOTATATE in the Treatment of Recurrent Pheochromocytoma With Multiple Metastases. Clin Nucl Med 2024; 49:338-339. [PMID: 38049973 DOI: 10.1097/rlu.0000000000004994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
ABSTRACT The treatment of metastatic pheochromocytoma is challenging. We report a case of a woman with recurrent pheochromocytoma with multiple metastases who achieved excellent response after 4 cycles of 177 Lu-DOTATATE therapy. She did not experience any observable adverse effects. Her disease was still stable 6 months after the fourth cycle of treatment.
Collapse
Affiliation(s)
- Dan Su
- From the Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province; and Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | | | | | | | | |
Collapse
|
17
|
Kornerup LS, Andreassen M, Knigge U, Arveschoug AK, Poulsen PL, Kjær A, Oturai PS, Grønbæk H, Dam G. Effects of Peptide Receptor Radiotherapy in Patients with Advanced Paraganglioma and Pheochromocytoma: A Nation-Wide Cohort Study. Cancers (Basel) 2024; 16:1349. [PMID: 38611027 PMCID: PMC11010872 DOI: 10.3390/cancers16071349] [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: 02/26/2024] [Revised: 03/16/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION Pheochromocytomas and paragangliomas are rare neuroendocrine tumours that originate from chromaffin cells within the adrenal medulla or extra-adrenal sympathetic ganglia. Management of disseminated or metastatic pheochromocytomas and paragangliomas continues to pose challenges and relies on limited evidence. METHOD In this study, we report retrospective data on median overall survival (OS) and median progression-free survival (PFS) for all Danish patients treated with peptide receptor radionuclide therapy (PRRT) with 177Lu-Dotatate or 90Y-Dotatate over the past 15 years. One standard treatment of PRRT consisted of 4 consecutive cycles with 8-14-week intervals. RESULTS We included 28 patients; 10 were diagnosed with pheochromocytoma and 18 with paraganglioma. Median age at first PRRT was 47 (IQR 15-76) years. The median follow-up time was 31 (IQR 17-37) months. Eight patients died during follow-up. Median OS was 72 months, and 5-year survival was 65% with no difference between pheochromocytoma and paraganglioma. Patients with germline mutations had better survival than patients without mutations (p = 0.041). Median PFS after the first cycle of PRRT was 30 months. For patients who previously received systemic treatment, the median PFS was 19 months, compared with 32 months for patients with no previous systemic treatment (p = 0.083). CONCLUSIONS The median OS of around 6 years and median PFS of around 2.5 years found in this study are comparable to those reported in previous studies employing PRRT. Based on historical data, the efficacy of PRRT may be superior to 131I-MIBG therapy, and targeted therapy with sunitinib and PRRT might therefore be considered as first-line treatment in this patient group.
Collapse
Affiliation(s)
- Linda Skibsted Kornerup
- Department of Hepatology & Gastroenterology, ENETS Center of Excellence, Aarhus University Hospital, 8200 Aarhus, Denmark; (L.S.K.); (H.G.); (G.D.)
| | - Mikkel Andreassen
- Department of Endocrinology, ENETS Center of Excellence, Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Ulrich Knigge
- Department of Endocrinology, ENETS Center of Excellence, Rigshospitalet, 2100 Copenhagen, Denmark;
- Department of Surgery and Transplantation, ENETS Center of Excellence, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Anne Kirstine Arveschoug
- Department of Nuclear Medicine & PET, ENETS Center of Excellence, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Per Løgstup Poulsen
- Department of Endocrinology, ENETS Center of Excellence, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Andreas Kjær
- Department of Clinical Physiology and Nuclear Medicine, ENETS Center of Excellence, Rigshospitalet, 2100 Copenhagen, Denmark; (A.K.); (P.S.O.)
| | - Peter Sandor Oturai
- Department of Clinical Physiology and Nuclear Medicine, ENETS Center of Excellence, Rigshospitalet, 2100 Copenhagen, Denmark; (A.K.); (P.S.O.)
| | - Henning Grønbæk
- Department of Hepatology & Gastroenterology, ENETS Center of Excellence, Aarhus University Hospital, 8200 Aarhus, Denmark; (L.S.K.); (H.G.); (G.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Gitte Dam
- Department of Hepatology & Gastroenterology, ENETS Center of Excellence, Aarhus University Hospital, 8200 Aarhus, Denmark; (L.S.K.); (H.G.); (G.D.)
| |
Collapse
|
18
|
Baudin E, Goichot B, Berruti A, Hadoux J, Moalla S, Laboureau S, Nölting S, de la Fouchardière C, Kienitz T, Deutschbein T, Zovato S, Amar L, Haissaguerre M, Timmers H, Niccoli P, Faggiano A, Angokai M, Lamartina L, Luca F, Cosentini D, Hahner S, Beuschlein F, Attard M, Texier M, Fassnacht M. Sunitinib for metastatic progressive phaeochromocytomas and paragangliomas: results from FIRSTMAPPP, an academic, multicentre, international, randomised, placebo-controlled, double-blind, phase 2 trial. Lancet 2024; 403:1061-1070. [PMID: 38402886 DOI: 10.1016/s0140-6736(23)02554-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/04/2023] [Accepted: 11/13/2023] [Indexed: 02/27/2024]
Abstract
BACKGROUND No randomised controlled trial has ever been done in patients with metastatic phaeochromocytomas and paragangliomas. Preclinical and first clinical evidence suggested beneficial effects of sunitinib. We aimed to evaluate the safety and efficacy of sunitinib in patients with metastatic phaeochromocytomas and paragangliomas. METHODS FIRSTMAPPP is a multicentre, international, randomised, placebo-controlled, double-blind, phase 2 trial done at 14 academic centres across four European countries. Eligible participants were adults (aged ≥18 years) with sporadic or inherited progressive metastatic phaeochromocytomas and paragangliomas. Patients were randomly assigned (1:1) to receive either oral sunitinib (37·5 mg per day) or placebo. Randomisation was stratified according to SDHB status (mutation present vs wild type) and number of previous systemic therapies (0 vs ≥1). Primary endpoint was the rate of progression-free survival at 12 months according to real-time central review (Response Evaluation Criteria in Solid Tumours version 1.1). On the basis of a two-step Simon model, we aimed for the accrual of 78 patients, assuming a 20% improvement of the 12-month progression-free survival rate from 20% to 40%, to conclude that sunitinib is effective. Crossover from the placebo group was allowed. This trial is registered with ClinicalTrials.gov, number NCT01371201, and is closed for enrolment. FINDINGS From Dec 1, 2011, to Jan 31, 2019, a total of 78 patients with progressive metastatic phaeochromocytomas and paragangliomas were enrolled (39 patients per group). 25 (32%) of 78 patients had germline SDHx variants and 54 (69%) had used previous therapies. The primary endpoint was met, with a 12-month progression-free survival in 14 of 39 patients (36% [90% CI 23-50]) in the sunitinib group. In the placebo group, the 12-month progression-free survival in seven of 39 patients was 19% (90% CI 11-31), validating the hypotheses of our study design. The most frequent grade 3 or 4 adverse events were asthenia (seven [18%] of 39 and one [3%] of 39), hypertension (five [13%] and four [10%]), and back or bone pain (one [3%] and three [8%]) in the sunitinib and placebo groups, respectively. Three deaths occurred in the sunitinib group: these deaths were due to respiratory insufficiency, amyotrophic lateral sclerosis, and rectal bleeding. Only the latter event was considered drug related. Two deaths occurred in the placebo group due to aspiration pneumonia and septic shock. INTERPRETATION This first randomised trial supports the use of sunitinib as the medical option with the highest level of evidence for anti-tumour efficacy in progressive metastatic phaeochromocytomas and paragangliomas. FUNDING French Ministry of Health, through the National Institute for Cancer, German Ministry of Education and Research, and the German Research Foundation within the CRC/Transregio 205/2, EU Seventh Framework Programme, and a private donator grant.
Collapse
Affiliation(s)
- Eric Baudin
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif, France.
| | - Bernard Goichot
- Department of Endocrinology, Hopital de Hautepierre-Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Azienda Ospedaliera Spedali Civili di Brescia, Brescia, Italy
| | - Julien Hadoux
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif, France
| | - Salma Moalla
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif, France
| | - Sandrine Laboureau
- Department of Endocrinology Diabetology Nutrition, Hopitaux Universitaires d'Angers, Angers, France
| | - Svenja Nölting
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | | | - Tina Kienitz
- Department of Endocrinology and Metabolism, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Timo Deutschbein
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Stefania Zovato
- Familial Cancer Clinics, Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Laurence Amar
- Department of Hypertension PARIS, Hopital Europeen Georges-Pompidou, Université Paris Cité, Paris, France
| | | | - Henri Timmers
- Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Patricia Niccoli
- Department of Medical Oncology, Institut Paoli Calmette, Marseille, France
| | - Antongiulio Faggiano
- Department of Clinical Medicine and Surgery, Endocrinology, Diabetology and Andrology Unit, Federico II University of Naples, Naples, Italy
| | - Moussa Angokai
- Office of Biostatistics and Epidemiology, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Livia Lamartina
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif, France
| | - Florina Luca
- Department of Endocrinology, Hopital de Hautepierre-Hopitaux Universitaires de Strasbourg, Strasbourg, France
| | - Deborah Cosentini
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Azienda Ospedaliera Spedali Civili di Brescia, Brescia, Italy
| | - Stefanie Hahner
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Marie Attard
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif, France
| | - Matthieu Texier
- Office of Biostatistics and Epidemiology, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Vukomanovic V, Nedic KV, Radojevic MZ, Dagovic A, Milosavljevic N, Markovic M, Ignjatovic V, Simic Vukomanovic I, Djukic S, Sreckovic M, Backovic M, Vuleta M, Djukic A, Vukicevic V, Ignjatovic V. Predicting the survival probability of functional neuroendocrine tumors treated with peptide receptor radionuclide therapy: Serbian experience. Front Endocrinol (Lausanne) 2024; 14:1270421. [PMID: 38317712 PMCID: PMC10840135 DOI: 10.3389/fendo.2023.1270421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/27/2023] [Indexed: 02/07/2024] Open
Abstract
Introduction Peptide receptor radionuclide therapy (PRRT) is a treatment option for well-differentiated, somatostatin receptor positive, unresectable or/and metastatic neuroendocrine tumors (NETs). Although high disease control rates seen with PRRT a significant number NET patients have a short progression-free interval, and currently, there is a deficiency of effective biomarkers to pre-identify these patients. This study is aimed at determining the prognostic significance of biomarkers on survival of patients with NETs in initial PRRT treatment. Methodology We retrospectively analyzed 51 patients with NETs treated with PRRT at the Department for nuclear medicine, University Clinical Center Kragujevac, Serbia, with a five-year follow-up. Eligible patients with confirmed inoperable NETs, were retrospectively evaluated hematological, blood-based inflammatory markers, biochemical markers and clinical characteristics on disease progression. In accordance with the progression og the disease, the patients were divided into two groups: progression group (n=18) and a non-progression group (n=33). Clinical data were compared between the two groups. Results A total of 51 patients (Md=60, age 25-75 years) were treated with PRRT, of whom 29 (56.86%) demonstrated stable disease, 4 (7.84%) demonstrated a partial response, and 14 (27.46%) demonstrated progressive disease and death was recorded in 4 (7.84%) patients. The mean PFS was a 36.22 months (95% CI 30.14-42.29) and the mean OS was 44.68 months (95% CI 37.40-51.97). Univariate logistic regression analysis displayed that age (p<0.05), functional tumors (p<0.05), absolute neutrophil count (p<0.05), neutrophil-lymphocyte ratio-NLR (p<0.05), C-reactive protein-CRP (p<0.05), CRP/Albumin (p<0.05), alanine aminotransferase-ALT (p<0.05), were risk factors for disease progression. Multivariate logistic regression analysis exhibited that functional tumors (p<0.001), age (p<0.05), CRP (p<0.05), and ALT (p<0.05), were independent risk factors for the disease progression in patients with NETs. Tumor functionality was the most powerful prognostic factor. The median PFS (11.86 ± 1.41 vs. 43.38 ± 3.16 months; p=0.001) and OS (21.81 ± 2.70 vs 53.86 ± 3.70, p=0.001) were significantly shorter in patients with functional than non-functional NETs respectively. Conclusion The study's results suggest that tumor functionality, and certain biomarkers may serve as prognostic survival indicators for patients with NETs undergoing PRRT. The findings can potentially help to identify patients who are at higher risk of disease progression and tailor treatment strategies accordingly.
Collapse
Affiliation(s)
- Vladimir Vukomanovic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Nuclear Medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Katarina Vuleta Nedic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Nuclear Medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marija Zivkovic Radojevic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Radiotherapy, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Aleksandar Dagovic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Medical Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Neda Milosavljevic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Radiotherapy, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Department for Medical Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladimir Ignjatovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Clinic for Cardiology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Ivana Simic Vukomanovic
- Department of Social Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Health Promotion, Institute of Public Health, Kragujevac, Serbia
| | - Svetlana Djukic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Clinic for Hematology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marijana Sreckovic
- Department of Medical and Business-Technological, Academy of Professional Studies Sabac, Sabac, Serbia
| | - Milena Backovic
- Department for Pathology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marko Vuleta
- Department for Cardiology, Clinical Hospital Center “Dr Dragisa Misovic Dedinje”, Belgrade, Serbia
| | - Aleksandar Djukic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center Kragujevac, Kragujevac, Serbia
| | | | - Vesna Ignjatovic
- Department of Nuclear Medicine and Clinical Oncology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department for Nuclear Medicine, University Clinical Center Kragujevac, Kragujevac, Serbia
| |
Collapse
|
20
|
Tang CYL, Chua WM, Huang HL, Lam WWC, Loh LM, Tai D, Ong SYK, Yan SX, Loke KSH, Ng DCE, Tham WY. Safety and efficacy of peptide receptor radionuclide therapy in patients with advanced pheochromocytoma and paraganglioma: A single-institution experience and review of the literature. J Neuroendocrinol 2023; 35:e13349. [PMID: 37937484 DOI: 10.1111/jne.13349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Despite advances in diagnosis and management, patients with advanced pheochromocytomas and paragangliomas (PPGL) face limited treatment options. This study aims to evaluate the safety and efficacy of peptide receptor radionuclide therapy (PRRT) in patients with advanced PPGL, based on a single-institution experience and provide a comprehensive review of the literature. METHODS A retrospective analysis was conducted on patients with advanced pheochromocytoma and paraganglioma who received PRRT at a single institution from April 2012 to March 2022. Clinical characteristics, treatment response, adverse events, and survival outcomes were assessed. A systematic literature review was also performed. RESULTS A total of 15 patients with advanced PPGL were included, the majority of whom had both metastatic and functional disease. Most patients received four infusions of 177Lu-DOTATATE (73%). The median therapeutic 177Lu-DOTATATE radioactivity for each infusion was 7.4 GBq. Only one patient was treated with one infusion of 90Y-DOTATATE (4.2 GBq) in addition to three infusions of Lu-177 DOTATATE. Overall, PRRT suggests a promising efficacy with disease control rate of 63.6% by RECIST v1.1. The median overall survival (OS) was not reached and the median progression free survival (PFS) was 25.9 months. In terms of safety, PRRT was well tolerated. Review of the literature revealed consistent findings, supporting the efficacy and safety of PRRT in PPGL. CONCLUSION This study suggests that PRRT is a safe and effective therapeutic option for patients with PPGL. Our findings align with the existing literature, providing additional evidence to support the use of PRRT in this challenging patient population.
Collapse
Affiliation(s)
- Charlene Yu Lin Tang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Wei Ming Chua
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Department of Neuroradiology, Singapore General Hospital, Singapore, Singapore
| | - Hian Liang Huang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Winnie Wing-Chuen Lam
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Lih Ming Loh
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - David Tai
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Simon Yew Kuang Ong
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Sean Xuexian Yan
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Kelvin S H Loke
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - David Chee-Eng Ng
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Wei Ying Tham
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
21
|
Fischer A, Kloos S, Remde H, Dischinger U, Pamporaki C, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Bechmann N, Hantel C, Mohr H, Pellegata NS, Bornstein SR, Kroiss M, Auernhammer CJ, Reincke M, Pacak K, Grossman AB, Beuschlein F, Nölting S. Responses to systemic therapy in metastatic pheochromocytoma/paraganglioma: a retrospective multicenter cohort study. Eur J Endocrinol 2023; 189:546-565. [PMID: 37949483 DOI: 10.1093/ejendo/lvad146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/02/2023] [Accepted: 09/19/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The therapeutic options for metastatic pheochromocytomas/paragangliomas (mPPGLs) include chemotherapy with cyclophosphamide/vincristine/dacarbazine (CVD), temozolomide monotherapy, radionuclide therapies, and tyrosine kinase inhibitors such as sunitinib. The objective of this multicenter retrospective study was to evaluate and compare the responses of mPPGLs including those with pathogenic variants in succinate dehydrogenase subunit B (SDHB), to different systemic treatments. DESIGN This is a retrospective analysis of treatment responses of mPPGL patients (n = 74) to systemic therapies. METHODS Patients with mPPGLs treated at 6 specialized national centers were selected based on participation in the ENSAT registry. Survival until detected progression (SDP) and disease-control rates (DCRs) at 3 months were evaluated based on imaging reports. RESULTS For the group of patients with progressive disease at baseline (83.8% of 74 patients), the DCR with first-line CVD chemotherapy was 75.0% (n = 4, SDP 11 months; SDHB [n = 1]: DCR 100%, SDP 30 months), with somatostatin peptide receptor-based radionuclide therapy (PPRT) 85.7% (n = 21, SDP 17 months; SDHB [n = 10]: DCR 100%, SDP 14 months), with 131I-meta-iodobenzylguanidine (131I-MIBG) 82.6% (n = 23, SDP 43 months; SDHB [n = 4]: DCR 100%, SDP 24 months), with sunitinib 100% (n = 7, SDP 18 months; SDHB [n = 3]: DCR 100%, SDP 18 months), and with somatostatin analogs 100% (n = 4, SDP not reached). The DCR with temozolomide as second-line therapy was 60.0% (n = 5, SDP 10 months; SDHB [n = 4]: DCR 75%, SDP 10 months). CONCLUSIONS We demonstrate in a real-life clinical setting that all current therapies show reasonable efficacy in preventing disease progression, and this is equally true for patients with germline SDHB mutations.
Collapse
Affiliation(s)
- Alessa Fischer
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Simon Kloos
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Hanna Remde
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ulrich Dischinger
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Institute de Salud Carlos III, Madrid, Spain
| | - Stephanie M J Fliedner
- First Department of Medicine, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Katharina Wang
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Julian Maurer
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Astrid Reul
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse, Dresden, Germany
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Stefan R Bornstein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christoph J Auernhammer
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Reincke
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Karel Pacak
- Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD, United States
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, United Kingdom
- NET Unit, ENETS Center of Excellence, Royal Free Hospital, London, United Kingdom
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
22
|
Fischer A, Kloos S, Maccio U, Friemel J, Remde H, Fassnacht M, Pamporaki C, Eisenhofer G, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Zitzmann K, Bechmann N, Žygienė G, Richter S, Hantel C, Vetter D, Lehmann K, Mohr H, Pellegata NS, Ullrich M, Pietzsch J, Ziegler CG, Bornstein SR, Kroiss M, Reincke M, Pacak K, Grossman AB, Beuschlein F, Nölting S. Metastatic Pheochromocytoma and Paraganglioma: Somatostatin Receptor 2 Expression, Genetics, and Therapeutic Responses. J Clin Endocrinol Metab 2023; 108:2676-2685. [PMID: 36946182 PMCID: PMC10505550 DOI: 10.1210/clinem/dgad166] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/01/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023]
Abstract
CONTEXT Pheochromocytomas and paragangliomas (PPGLs) with pathogenic mutations in the succinate dehydrogenase subunit B (SDHB) are associated with a high metastatic risk. Somatostatin receptor 2 (SSTR2)-dependent imaging is the most sensitive imaging modality for SDHB-related PPGLs, suggesting that SSTR2 expression is a significant cell surface therapeutic biomarker of such tumors. OBJECTIVE Exploration of the relationship between SSTR2 immunoreactivity and SDHB immunoreactivity, mutational status, and clinical behavior of PPGLs. Evaluation of SSTR-based therapies in metastatic PPGLs. METHODS Retrospective analysis of a multicenter cohort of PPGLs at 6 specialized Endocrine Tumor Centers in Germany, The Netherlands, and Switzerland. Patients with PPGLs participating in the ENSAT registry were included. Clinical data were extracted from medical records, and immunohistochemistry (IHC) for SDHB and SSTR2 was performed in patients with available tumor tissue. Immunoreactivity of SSTR2 was investigated using Volante scores. The main outcome measure was the association of SSTR2 IHC positivity with genetic and clinical-pathological features of PPGLs. RESULTS Of 202 patients with PPGLs, 50% were SSTR2 positive. SSTR2 positivity was significantly associated with SDHB- and SDHx-related PPGLs, with the strongest SSTR2 staining intensity in SDHB-related PPGLs (P = .01). Moreover, SSTR2 expression was significantly associated with metastatic disease independent of SDHB/SDHx mutation status (P < .001). In metastatic PPGLs, the disease control rate with first-line SSTR-based radionuclide therapy was 67% (n = 22, n = 11 SDHx), and with first-line "cold" somatostatin analogs 100% (n = 6, n = 3 SDHx). CONCLUSION SSTR2 expression was independently associated with SDHB/SDHx mutations and metastatic disease. We confirm a high disease control rate of somatostatin receptor-based therapies in metastatic PPGLs.
Collapse
Affiliation(s)
- Alessa Fischer
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
| | - Simon Kloos
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
| | - Umberto Maccio
- Department of Pathology and Molecular Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Juliane Friemel
- Department of Pathology and Molecular Pathology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Hanna Remde
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Graeme Eisenhofer
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Henri J L M Timmers
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - Stephanie M J Fliedner
- First Department of Medicine, University Medical Center Schleswig-Holstein, 23538 Lübeck, Germany
| | - Katharina Wang
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Julian Maurer
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Astrid Reul
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
| | - Kathrin Zitzmann
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse, 01307 Dresden, Germany
| | - Gintarė Žygienė
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse, 01307 Dresden, Germany
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse, 01307 Dresden, Germany
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Diana Vetter
- Department of Visceral and Transplantation Surgery, University Hospital, 8091 Zürich, Switzerland
| | - Kuno Lehmann
- Department of Visceral and Transplantation Surgery, University Hospital, 8091 Zürich, Switzerland
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Christian G Ziegler
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stefan R Bornstein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Martin Reincke
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, MD 20847, USA
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, UK
- NET Unit, ENETS Centre of Excellence, Royal Free Hospital, London, UK
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), CH-8091 Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| |
Collapse
|
23
|
Su D, Yang H, Qiu C, Chen Y. Peptide receptor radionuclide therapy in advanced Pheochromocytomas and Paragangliomas: a systematic review and meta-analysis. Front Oncol 2023; 13:1141648. [PMID: 37483516 PMCID: PMC10358840 DOI: 10.3389/fonc.2023.1141648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Objective Peptide receptor radionuclide therapy (PRRT) for advanced pheochromocytomas and paragangliomas (PPGLs) has received increasing attention. The purpose of this article is to evaluate the efficacy and safety of PRRT in patients with metastatic or inoperable PPGLs by meta-analysis. Methods A literature search was conducted in PubMed, Embase, Scopus, and Cochrane Library databases up to November 2022. All articles on PRRT for PPGLs were searched, and appropriate data were included for analysis. The measures evaluated included objective response rate (ORR), disease control rate (DCR), clinical response rate, biochemical response rate, progression-free survival (PFS), overall survival (OS), and adverse events. Statistical analysis was performed using Stata 16.0 and the R programming language, data were combined using a random-effects model, and the results were presented using forest plots. Results A total of 20 studies with 330 patients were included in the analysis. The results showed that ORR and DCR were 20.0% (95% CI: 12.0%-28.0%) and 90.0% (95% CI: 85.0%-95.0%), respectively. Clinical and biochemical responses were 74.9% (95% CI: 56.3%-90.2%) and 69.5% (95%CI: 40.2%-92.9%). Median PFS and median OS were 31.79 (95% CI:21.25-42.33) months and 74.30 (95% CI: 0.75-147.84) months, respectively. Any grade of hematotoxicity and nephrotoxicity occurred in 22.3% (95% CI:12.5%-33.5%) and 4.3% (95% CI:0.2%-11.4%) patients. Grade 3-4 hemotoxicity occurred in 4.3% (95% CI:0.2%-11.4%) and grade 3-4 nephrotoxicity in 4/212 patients. Additionally, Treatment was discontinued in 9.0% (95% CI: 0.5%-23.3%) patients and one patient died as a result of a toxicity. Conclusion Patients with metastatic or inoperable PPGLs can be effectively treated with PRRT, and it has a favorable safety profile. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42022359232.
Collapse
Affiliation(s)
- Dan Su
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Hongyu Yang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Chen Qiu
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
- Academician (expert) Workstation of Sichuan Province, Luzhou, Sichuan, China
| |
Collapse
|
24
|
Ren K, Feng J, Yang X, Ren B, Guo Y. Efficacy of metoprolol combined with torasemide in elderly patients with degenerative valvular heart disease and heart failure and its influence on NT-proBNP levels. Pak J Med Sci 2023; 39:945-949. [PMID: 37492298 PMCID: PMC10364288 DOI: 10.12669/pjms.39.4.7465] [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: 12/10/2022] [Revised: 12/23/2022] [Accepted: 05/20/2023] [Indexed: 07/27/2023] Open
Abstract
Objective To investigate the efficacy of metoprolol combined with torasemide in elderly patients with degenerative valvular heart disease (DVHD) and heart failure and its influence on N-terminal pro-B-type natriuretic peptide (NT-proBNP) concentrations. Methods The records of 129 DVHD patients ≥60 years old with heart failure diagnosed and treated in our hospital from January 2020 to February 2022 were retrospectively selected. According to the treatment records, 62 patients received metoprolol treatment (Control-group), and 67 patients received metoprolol combined with torasemide treatment (Observation-group). The changes in cardiac function, NT-proBNP and inflammatory factor concentrations and clinical efficacy were analyzed and compared between the two groups before and after treatment. Results After treatment, left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter (LVEDD) and the mitral ratio of peak early to late diastolic filling velocity (E/A) were lower, with a greater decrease in the Observation-group compared to the Control-group (P<0.05). After treatment, NT-proBNP and interleukin-1beta (IL-1β), tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6) concentrations were lower in both groups, with a greater decrease in the Observation-group compared to the Control-group (P<0.05). The total clinical efficacy of the Observation-group was significantly higher than the Control-group (P<0.05). There was no significant difference in the total incidence of adverse drug reactions between the two groups (P>0.05). Conclusion Metoprolol combined with torasemide has a significant therapeutic effect on DVHD patients with heart failure. This drug combination improved cardiac function, reduced NT-proBNP concentrations and has good safety.
Collapse
Affiliation(s)
- Kai Ren
- Kai Ren Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Jinping Feng
- Jinping Feng Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Xuwen Yang
- Xuwen Yang Department of Recovery Unit, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Bei Ren
- Bei Ren Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Yujun Guo
- Yujun Guo Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| |
Collapse
|
25
|
Zhang X, Wakabayashi H, Hiromasa T, Kayano D, Kinuya S. Recent Advances in Radiopharmaceutical Theranostics of Pheochromocytoma and Paraganglioma. Semin Nucl Med 2023; 53:503-516. [PMID: 36641337 DOI: 10.1053/j.semnuclmed.2022.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
As a rare kind of non-epithelial neuroendocrine neoplasms, paragangliomas (PGLs) exhibit various clinical characteristics with excessive catecholamine secretion and have been a research focus in recent years. Although several modalities are available nowadays, radiopharmaceuticals play an integral role in the management of PGLs. Theranostics utilises radiopharmaceuticals for diagnostic and therapeutic intentions by aiming at a specific target in tumour and has been considered a possible means in diagnosis, staging, monitoring and treatment planning. Numerous radiopharmaceuticals have been developed over the past decades. 123/131-Metaiodobenzylguanidine (123/131I-MIBG), the theranostics pair target on norepinephrine transporter system, has remained a fantastic protocol for patients with PGLs because of disease control with limited toxicity. The high-specific-activity 131I-MIBG was authorised by the Food and Drug Administration as a systemic treatment method for metastatic PGLs in 2018. Afterward, peptide receptor radionuclide therapy, which uses radiolabelled somatostatin (SST) analogues, has been exploited as a superior substitute. 68Ga-somatostatin analogue (SSA) PET showed significant performance in diagnosing PGLs than MIBG scintigraphy, especially in patients with head and neck PGLs or SDHx mutation. 90Y/177Lu-DOTA-SSA is highly successful and has preserved favourable safety with mounting evidence regarding objective response, disease stabilisation, symptomatic and hormonal management and quality of life preservation. Besides the ordinary beta emitters, alpha-emitters such as 211At-MABG and 225Ac-DOTATATE have been investigated intensively in recent years. However, many studies are still in the pre-clinical stage, and more research is necessary. This review summarises the developments and recent advances in radiopharmaceutical theranostics of PGLs.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.
| | - Tomo Hiromasa
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| |
Collapse
|
26
|
Marques P. The Effects of Peptide Receptor Radionuclide Therapy on the Neoplastic and Normal Pituitary. Cancers (Basel) 2023; 15:2710. [PMID: 37345047 PMCID: PMC10216433 DOI: 10.3390/cancers15102710] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
Pituitary neuroendocrine tumours (PitNETs) are usually benign and slow-growing; however, in some cases, they may behave aggressively and become resistant to conventional treatments. Therapeutic options for aggressive or metastatic PitNETs are limited, and currently mainly consist of temozolomide, with little experience of other emerging approaches, including peptide receptor radionuclide therapy (PRRT). Somatostatin receptor expression in PitNETs explains the effectiveness of somatostatin analogues for treating PitNETs, particularly those hypersecreting pituitary hormones, such as growth hormone or adrenocorticotropic hormone. The expression of such receptors in pituitary tumour cells has provided the rationale for using PRRT to treat patients with aggressive or metastatic PitNETs. However, the PRRT efficacy in this setting remains unestablished, as knowledge on this today is based only on few case reports and small series of cases, which are reviewed here. A total of 30 PRRT-treated patients have been thus far reported: 23 aggressive PitNETs, 5 carcinomas, and 2 of malignancy status unspecified. Of the 27 published cases with information regarding the response to PRRT, 5 (18%) showed a partial response, 8 (30%) had stable disease, and 14 (52%) had progressive disease. No major adverse effects have been reported, and there is also no increased risk of clinically relevant hypopituitarism in patients with pituitary or non-pituitary neuroendocrine tumours following PRRT. PRRT may be regarded as a safe option for patients with aggressive or metastatic PitNETs if other treatment approaches are not feasible or have failed in controlling the disease progression, with tumour shrinkage occurring in up to a fifth of cases, while about a third of aggressive pituitary tumours may achieve stable disease. Here, the data on PRRT in the management of patients with aggressive pituitary tumours are reviewed, as well as the effects of PRRT on the pituitary function in other PRRT-treated cancer patients.
Collapse
Affiliation(s)
- Pedro Marques
- Pituitary Tumor Unit, Endocrinology Department, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal;
- Faculdade de Medicina, Universidade Católica Portuguesa, 2635-631 Lisbon, Portugal
| |
Collapse
|
27
|
Urso L, Nieri A, Uccelli L, Castello A, Artioli P, Cittanti C, Marzola MC, Florimonte L, Castellani M, Bissoli S, Porto F, Boschi A, Evangelista L, Bartolomei M. Lutathera® Orphans: State of the Art and Future Application of Radioligand Therapy with 177Lu-DOTATATE. Pharmaceutics 2023; 15:pharmaceutics15041110. [PMID: 37111596 PMCID: PMC10142322 DOI: 10.3390/pharmaceutics15041110] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Lutathera® is the first EMA- and FDA-approved radiopharmaceutical for radioligand therapy (RLT). Currently, on the legacy of the NETTER1 trial, only adult patients with progressive unresectable somatostatin receptor (SSTR) positive gastroenteropancreatic (GEP) neuroendocrine neoplasms (NET) can be treated with Lutathera®. Conversely, patients with SSTR-positive disease arising from outside the gastroenteric region do not currently have access to Lutathera® treatment despite several papers in the literature reporting the effectiveness and safety of RLT in these settings. Moreover, patients with well-differentiated G3 GEP-NET are also still “Lutathera orphans”, and retreatment with RLT in patients with disease relapse is currently not approved. The aim of this critical review is to summarize current literature evidence assessing the role of Lutathera® outside the approved indications. Moreover, ongoing clinical trials evaluating new possible applications of Lutathera® will be considered and discussed to provide an updated picture of future investigations.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Licia Uccelli
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
- Correspondence: ; Tel.: +39-053-232-6387
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Paolo Artioli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Maria Cristina Marzola
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Luigia Florimonte
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Sergio Bissoli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Francesca Porto
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Laura Evangelista
- Department of Medicine DIMED, University of Padua, 35128 Padua, Italy;
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| |
Collapse
|
28
|
Marretta AL, Ottaiano A, Iervolino D, Bracigliano A, Clemente O, Di Gennaro F, Tafuto R, Santorsola M, Lastoria S, Tafuto S. Response to Peptide Receptor Radionuclide Therapy in Pheocromocytomas and Paragangliomas: A Systematic Review and Meta-Analysis. J Clin Med 2023; 12:jcm12041494. [PMID: 36836029 PMCID: PMC9964778 DOI: 10.3390/jcm12041494] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/03/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
INTRODUCTION Peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE and 90Y-DOTATOC showed efficacy in the metastatic setting of pheocromocytomas (PCCs) and paragangliomas (PGLs) where no standard therapies have been established. BACKGROUND A search of peer-reviewed and English articles reporting on 177Lu-DOTATATE and 90Y-DOTATOC efficacy was performed through Medline and Scopus. A subsequent meta-analysis was performed to evaluate the pooled effect size on disease control rate (DCR) with PRRT. Secondary endpoints were description of patients' genetic characteristics, hematologic toxicity, and time-to-outcome. The pooled effect was estimated with both a mixed-effects model and a random-effects model. RESULTS Twelve studies met the criteria for this meta-analysis: ten with 177Lu- and two with 90Y-PRRTs (213 patients). The largest one included 46 patients. Median ages ranged from 32.5 to 60.4 years. When reported, mutations of SDHB were the most frequent genetic alterations. The pooled DCRs were 0.83 (95% CI: 0.75-0.88) and 0.76 (95% CI: 0.56-0.89) for 177Lu- and 90Y-PRRT, respectively. The pooled DCR for PRRT was 0.81 (95% CI: 0.74-0.87). CONCLUSIONS We report an updated and solid estimate of DCR achieved with 177Lu- and 90Y-PRRT in PCCs and PGLs, showing that these therapies can be considered in the multidisciplinary treatment of PCCs and PGLs as alternatives to I-131 MIBG and chemotherapy.
Collapse
Affiliation(s)
- Antonella Lucia Marretta
- Department of Clinical and Surgery Oncology Unit, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Domenico Iervolino
- Pathology Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Alessandra Bracigliano
- Nuclear Medicine Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Ottavia Clemente
- Sarcomas and Rare Tumours Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-329-9786209
| | - Francesca Di Gennaro
- Nuclear Medicine Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Roberto Tafuto
- Department of Neuroscience and Reproductive and Dental Sciences, Division of Neurosurgery, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
| | - Mariachiara Santorsola
- SSD Innovative Therapies for Abdominal Metastases, Department of Abdominal Oncology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| | - Salvatore Tafuto
- Sarcomas and Rare Tumours Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy
| |
Collapse
|
29
|
Comparative evaluation of radionuclide therapy using 90Y and 177Lu. Ann Nucl Med 2023; 37:52-59. [PMID: 36352185 DOI: 10.1007/s12149-022-01803-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Both 90Y and 177Lu are attractive β-emitters for radionuclide therapy and have been used in clinical practice. Nevertheless, comparative evaluation between 90Y- and 177Lu-labeled molecules has not been fully conducted. Thus, in this study, the features of 90Y and 177Lu for radionuclide therapy were assessed in tumor-bearing mice. METHODS Two tumor cell lines with different growth rates were used. Biodistribution studies of 177Lu-labeled antibodies (177Lu-Abs) were conducted in each tumor-bearing mouse model. Subsequently, the therapeutic effect of 90Y- and 177Lu-Ab were assessed in tumor-bearing mice. The absorbed radiation dose for the tumor was estimated using the Monte Carlo simulation. RESULTS 177Lu-Abs demonstrated high tumor accumulation in both tumor-xerograph. In the fast-growing tumor model, 90Y-Ab showed a better therapeutic effect than 177Lu-Ab, reflecting a higher absorbed radiation dose of 90Y-Ab than that of 177Lu-Ab. In the slow-growing tumor model, both 90Y- and 177Lu-Ab showed an excellent therapeutic effect; however, 177Lu-Ab had a longer efficacy period than 90Y-Ab, which could be attributed to the longer half-life and better dose uniformity of 177Lu than those of 90Y. CONCLUSIONS To accomplish a maximum therapeutic effect, selecting 90Y or 177Lu, to depend on the growth rate of individual cancer, would be helpful.
Collapse
|
30
|
Advances in Endocrine Surgery. Surg Oncol Clin N Am 2023; 32:199-220. [PMID: 36410918 DOI: 10.1016/j.soc.2022.08.004] [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: 11/06/2022]
Abstract
Recent changes in the landscape of endocrine surgery include a shift from total thyroidectomy for almost all patients with papillary thyroid cancer to the incorporation of thyroid lobectomy for well-selected patients with low-risk disease; minimally invasive parathyroidectomy with, and potentially without, intraoperative parathyroid hormone monitoring for patients with well-localized primary hyperparathyroidism; improvement in the management of parathyroid cancer with the incorporation of immune checkpoint blockade and/or targeted therapies; and the incorporation of minimally invasive techniques in the management of patients with benign tumors and selected secondary malignancies of the adrenal gland.
Collapse
|
31
|
Araujo-Castro M, Pascual-Corrales E, Alonso-Gordoa T, Molina-Cerrillo J, Martínez Lorca A. Papel de las pruebas de imagen con radionúclidos en el diagnóstico y tratamiento de los feocromocitomas y paragangliomas. ENDOCRINOL DIAB NUTR 2022. [DOI: 10.1016/j.endinu.2021.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Araujo-Castro M, Pascual-Corrales E, Alonso-Gordoa T, Molina-Cerrillo J, Martínez Lorca A. Role of imaging test with radionuclides in the diagnosis and treatment of pheochromocytomas and paragangliomas. ENDOCRINOL DIAB NUTR 2022; 69:614-628. [PMID: 36402734 DOI: 10.1016/j.endien.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/29/2021] [Indexed: 06/16/2023]
Abstract
Radionuclide imaging tests with [123I] Metaiodobenzylguanidine (MIBG), [18F] -fluorodeoxyglucose, [18F]-fluorodopa, or 68Ga-DOTA(0)-Tyr(3)-octreotate are useful for the diagnosis, staging and follow-up of pheochromocytomas (PHEOs) and paragangliomas (PGLs) (PPGLs). In addition to their ability to detect and localize the disease, they allow a better molecular characterization of the tumours, which is useful for planning targeted therapy with iodine-131 (131I) -labelled MIBG or with peptide receptor radionuclide therapy (PRRT) with [177Lu]-labelled DOTATATE or other related agents in patients with metastatic disease. In this review we detail the main characteristics of the radiopharmaceuticals used in the functional study of PPGLs and the role of nuclear medicine tests for initial evaluation, staging, selection of patients for targeted molecular therapy, and radiation therapy planning. It also offers a series of practical recommendations regarding the functional imaging according to the different clinical and genetic scenarios in which PPGLs occur, and on the indications and efficacy of therapy with [131I]-MIBG and 177Lu-DOTATATE.
Collapse
Affiliation(s)
- Marta Araujo-Castro
- Unidad de Neuroendocrinología, Servicio de Endocrinología y Nutrición, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain; Universidad de Alcalá, Departamento de Ciencias de la Salud, Madrid, Spain.
| | - Eider Pascual-Corrales
- Unidad de Neuroendocrinología, Servicio de Endocrinología y Nutrición, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Teresa Alonso-Gordoa
- Servicio de Oncología Médica, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Molina-Cerrillo
- Servicio de Oncología Médica, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alberto Martínez Lorca
- Servicio de Medicina Nuclear, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain.
| |
Collapse
|
33
|
Sobocki BK, Perdyan A, Szot O, Rutkowski J. Management of Pheochromocytomas and Paragangliomas: A Case-Based Review of Clinical Aspects and Perspectives. J Clin Med 2022; 11:jcm11092591. [PMID: 35566714 PMCID: PMC9103340 DOI: 10.3390/jcm11092591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 12/12/2022] Open
Abstract
Paraganglioma and pheochromocytoma are rare medical conditions. Thus, there are still a small number of studies, clinical trials, and evidence-based data in this field. This makes clinical decisions more difficult. In this study, we present a case report enriched with a short review of available essential clinical data, indicating the need for constant metoxycatecholamine level observation and a proper diagnostic imaging approach, especially in terms of ongoing pandemics. Our research also provides a summary of the molecular background of these diseases, indicating their future role in clinical management. We analyzed the ClinicalTrials.gov dataset in order to show future perspectives. In this paper, the use of the PET-CT before MRI or CT is proposed in specific cases during diagnosis processes contrary to the guidelines. PET-CT may be as effective as standard procedures and may provide a faster diagnosis, which is important in periods with more difficult access to health care, such as during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Bartosz Kamil Sobocki
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-214 Gdansk, Poland;
- Correspondence: (B.K.S.); (J.R.)
| | - Adrian Perdyan
- International Research Agenda 3P Medicine Laboratory, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Olga Szot
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Jacek Rutkowski
- Department of Oncology and Radiotherapy, Medical University of Gdansk, 80-214 Gdansk, Poland
- Correspondence: (B.K.S.); (J.R.)
| |
Collapse
|
34
|
Wang P, Hou G, Jing H, Zhuang H, Li F. Similar Findings on 18F-MFBG PET/CT and 68Ga-DOTATATE PET/CT in a Patient With Widespread Metastatic Pheochromocytoma. Clin Nucl Med 2022; 47:451-453. [PMID: 35020650 DOI: 10.1097/rlu.0000000000003997] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT A 27-year-old woman with known malignant pheochromocytoma was enrolled in a clinical trial to compare efficacy of 68Ga-DOTATATE PET/CT and 18F-MFBG PET/CT. Images from both studies detected similar number of the metastatic lesions. In addition, both studies detected more lesions than 131I-MIBG imaging.
Collapse
Affiliation(s)
| | | | | | - Hongming Zhuang
- Department of Radiology, Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | |
Collapse
|
35
|
Puliani G, Chiefari A, Mormando M, Bianchini M, Lauretta R, Appetecchia M. New Insights in PRRT: Lessons From 2021. Front Endocrinol (Lausanne) 2022; 13:861434. [PMID: 35450421 PMCID: PMC9016202 DOI: 10.3389/fendo.2022.861434] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using radiolabeled somatostatin analogs has been used for over two decades for the treatment of well-differentiated neuroendocrine tumors (NETs), and the publication of the NETTER-1 trials has further strengthened its clinical use. However, many aspects of this treatment are still under discussion. The purpose of this review is to collect and discuss the new available evidence, published in 2021, on the use of 177Lu-Oxodotreotide (DOTATATE) or 90Y-Edotreotide (DOTATOC) in adult patients with NETs focusing on the following hot topics: 1) PRRT use in new clinical settings, broaden its indications; 2) the short- and long-term safety; and 3) the identification of prognostic and predictive factors. The review suggests a possible future increase of PRRT applications, using it in other NETs, as a neoadjuvant treatment, or for rechallenge. Regarding safety, available studies, even those with long follow-up, supported the low rates of adverse events, even though 1.8% of treated patients developed a second malignancy. Finally, there is a lack of prognostic and predictive factors for PRRT, with the exception of the crucial role of nuclear imaging for both patient selection and treatment response estimation.
Collapse
Affiliation(s)
- Giulia Puliani
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Alfonsina Chiefari
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marilda Mormando
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marta Bianchini
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Rosa Lauretta
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
36
|
Yadav MP, Ballal S, Sahoo RK, Bal C. Efficacy and safety of 225Ac-DOTATATE targeted alpha therapy in metastatic paragangliomas: a pilot study. Eur J Nucl Med Mol Imaging 2022; 49:1595-1606. [PMID: 34837103 PMCID: PMC8626283 DOI: 10.1007/s00259-021-05632-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/20/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE In this study, we aim to evaluate the efficacy and safety of 225AC-DOTATATE targeted alpha therapy in advanced-stage paragangliomas (PGLs). METHODS Nine (6 males and 3 females) consecutive patients with histologically proven PGLs were treated with 225Ac-DOTATATE targeted alpha therapy (TAT) and concomitant radiosensitizer, capecitabine, at 8-weekly intervals up to a cumulative activity of ~ 74 MBq. The primary endpoint included evaluating therapy response and disease control rate (DCR) using the RECIST 1.1 criteria. Additional secondary endpoints comprised clinical response assessment using EORTC QLQ-H&N35 questionnaire, Karnofsky Performance Scale (KPS), Eastern Cooperative Oncology Group performance status (ECOG), analgesic score (AS), dose alterations of anti-hypertensive drugs (anti-HTN), and the safety and side-effect profile evaluation as per CTCAE criteria version 5.0. RESULTS Following 225Ac-DOTATATE treatment, morphological response revealed partial response in 50%, stable disease in 37.5%, and disease progression in 12.5%, with a DCR of 87.5%. Similarly, the symptomatic response was remarkable, and anti-HTN drugs were stopped in 25% and reduced in 37.5%. Another significant finding in our study revealed a morphologic DCR of 66.6% (2/3) in patients who failed previous lutetium-177 peptide receptor radionuclide therapy (177Lu-PRRT). Regarding the KPS, ECOG, and AS performance scores, a notable improvement was observed post-225Ac-DOTATATE treatment. The QLQ-H&N35 symptom scores evaluated in seven H&N PGL patients showed significant improvement in all aspects. No improvement in sexual function was noted (P = 0.3559). Despite the significant reduction in the analgesic score post-treatment (P = 0.0031), the QLQ-H&N35 revealed only marginal significance concerning the intake of pain killers (P = 0.1723). No grade III/IV hematological, renal, and hepatological toxicities were noted. CONCLUSION The evidence from this study suggests 225Ac-DOTATATE therapy is effective and safe in the treatment of advanced-stage PGLs and also reports a clear benefit even in patient's refractory to the previous 177Lu-PRRT.
Collapse
Affiliation(s)
- Madhav Prasad Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, BR Ambedkar Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
37
|
New Directions in Treatment of Metastatic or Advanced Pheochromocytomas and Sympathetic Paragangliomas: an American, Contemporary, Pragmatic Approach. Curr Oncol Rep 2022; 24:89-98. [DOI: 10.1007/s11912-022-01197-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
|
38
|
Marini I, Sansovini M, Bongiovanni A, Nicolini S, Grassi I, Ranallo N, Monti M, DI Iorio V, Germanò L, Caroli P, Sarnelli A, Paganelli G, Severi S. Theragnostic in neuroendocrine tumors. 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... 2021; 65:342-352. [PMID: 34881852 DOI: 10.23736/s1824-4785.21.03426-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In the last few decades, the incidence and prevalence of neuroendocrine tumors has been increasing. The theragnostic approach, that allows the diagnosis and treatment of different neoplasms with the same ligand, is a typical nuclear medicine tool. Applied for years, is also pivotal in neuroendocrine tumors (NETs) where it has improved the diagnostic accuracy and the therapeutic efficacy with impact on patient's survival. Theragnostic also allows the identification of important prognostic factors such as tumor location and burden, presence of liver metastases and intensity of somatostatin receptors (SSTR) expression to consider in new and possibly combined studies to ameliorate patient's outcome. Moreover, the possibility to evaluate receptor expression even in non-NET malignancies has de facto widened the possible indications for PRRT. We believe that this innovative therapeutic approach will be implemented in next years by radiomics and biological tumors characterization to better address PRRT applications.
Collapse
Affiliation(s)
- Irene Marini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Maddalena Sansovini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Silvia Nicolini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Ilaria Grassi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumors Center - CDO-TR, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Manuela Monti
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Valentina DI Iorio
- Unit of Oncological Pharmacy, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Luca Germanò
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Paola Caroli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Anna Sarnelli
- Unit of Medical Physics, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Giovanni Paganelli
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy
| | - Stefano Severi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori - IRST Dino Amadori, Meldola, Forlì-Cesena, Italy -
| |
Collapse
|
39
|
Special situations in pheochromocytomas and paragangliomas: pregnancy, metastatic disease, and cyanotic congenital heart diseases. Clin Exp Med 2021; 22:359-370. [PMID: 34591219 DOI: 10.1007/s10238-021-00763-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/18/2021] [Indexed: 01/08/2023]
Abstract
The aim of our study was to describe the epidemiology, diagnosis, and treatment of the most complex pheochromocytoma and paraganglioma (PGL) cases, including pheochromocytoma/PGL during pregnancy, in cyanotic congenital heart diseases (CCHDs), and metastatic pheochromocytoma. The English and Spanish literature was thoroughly evaluated searching for articles reporting clinical studies, case reports, or reviews of pheochromocytoma/PGL in pregnancy and in CCHD and metastatic pheochromocytoma/PGL. Particular settings in the diagnosis and management of pheochromocytoma and PGLs remain challenging. Those special situations include the diagnosis during pregnancy or in the context of CCHD since the typical clinical features of pheochromocytoma may be confounded with preeclampsia during pregnancy and with the complications commonly observed in CCHD. In addition, although some clinical and genetic features have been associated with higher risk of metastatic pheochromocytoma, the detection and prediction of the development of metastatic disease involve another complex situation that may require special hormonal determinations as plasmatic 3-methoxytyramine and nuclear medicine studies including 18FDG PET-CT or 18F-FDOPA PET-CT, among others. Furthermore, the selection of the most appropriate treatment in these situations, as well as the moment to carry it out, requires special care as limited evidence is available. This article reviews the epidemiology, diagnosis, and treatment of the pheochromocytoma/PGL during pregnancy, metastatic pheochromocytoma/PGL, and pheochromocytoma/PGL in CCHD. The diagnosis, and especially the treatment, of metastatic pheochromocytomas and pheochromocytoma/PGL during pregnancy and in CCHD is challenging. Thus, these cases should be management in reference centres by multidisciplinary teams specialized in the pheochromocytoma/PGL treatment.
Collapse
|