1
|
Kumar SA, Selvaraj VK, Srivastava MK. Circlet Sign: Urinary Contamination on 177Lu-DOTATATE Posttherapy Imaging. Clin Nucl Med 2025:00003072-990000000-01736. [PMID: 40392124 DOI: 10.1097/rlu.0000000000005990] [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: 02/27/2025] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
Peptide receptor radionuclide therapy has revolutionized the management of somatostatin receptor-positive metastatic neuroendocrine tumours. Treating nuclear medicine physicians must be aware of the radiation hazards posed by radioactive contamination and the imaging fallacies attributable to radioactive contamination on posttherapy planar imaging. The image fallacies, if not properly looked upon, upstage the disease. Also, radioactive contamination with body fluids on any external structure in close contact with the patients poses a prolonged radiation hazard. The authors in this present research demonstrate image fallacy due to radioactive urinary contamination on 177Lu-DOTATATE posttherapy imaging.
Collapse
Affiliation(s)
- Srinivas Ananth Kumar
- Department of Nuclear Medicine, Nizam's Institute of Medical Sciences, Hyderabad, TG, India
| | | | | |
Collapse
|
2
|
Mititelu MR, Stanulovic V, Mitoi A, Bucurica S, Hodolic M, Kairemo K. Overview of Adverse Reactions of Radiopharmaceuticals. Clin Oncol (R Coll Radiol) 2025:103857. [PMID: 40413089 DOI: 10.1016/j.clon.2025.103857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025]
Abstract
AIMS Theranostics utilizes the nuclear properties of radioactive isotopes, especially for molecular imaging and targeted therapy. Radiopharmaceuticals (RPs), which combine a pharmaceutical ligand with a radionuclide, enable accurate diagnosis and treatment of various diseases through modalities such as PET and SPECT imaging. The aim of this papare is to review adverse reactions associated with diagnostic and therapeutic radiopharmaceuticals, with an emphasis on their severity and clinical management. MATERIALS AND METHODS This review evaluates documented adverse effects (AEs) related to RPs used in nuclear medicine imaging (PET and SPECT) and radionuclide therapy, focusing on their severity and clinical management strategies. It also considers the mechanisms of RPs toxicity, distinguishes between general and specific AEs, and highlights the limitations in current adverse drug reaction (ADR) assessment tools. The methodology used was the research and synthesis of most relevant published literature data; most relevant papers were synthesized regarding the reporting system of ARs and categorized by the specific and systemic adverse effects of RPs. RESULTS Side effects from diagnostic RPs are relatively rare and typically minimal. Therapeutic RPs, selected for their high-energy radiation properties, can cause DNA damage to malignant cells while minimizing harm to healthy tissues. Although adverse effects do occur, they are generally fewer and less severe compared to conventional therapies. Severe toxicity is rare and often preventable. Both patient- and provider-reported ADRs offer important safety insights, though validated assessment instruments remain limited. CONCLUSION Radionuclide therapy offers a targeted approach that is a less invasive alternative to conventional treatments with a favorable safety profile. Continued evaluation of adverse reactions and the development of standardized ADR assessment tools are essential for improving patient outcomes and RP safety monitoring.
Collapse
Affiliation(s)
- M R Mititelu
- University of Medicine and Pharmacy Carol Davila, Clinic of Nuclear Medicine, Central University Emergency Military Hospital, Bucharest, Romania.
| | - V Stanulovic
- Patient Safety &Pharmacovigilance, Sanofi, Lyon, France.
| | - A Mitoi
- University of Medicine and Pharmacy Carol Davila, Clinic of Nuclear Medicine, Central University Emergency Military Hospital, Bucharest, Romania.
| | - S Bucurica
- University of Medicine and Pharmacy Carol Davila, Department of Gastroenterology, Central University Emergency Military Hospital, Bucharest, Romania.
| | - M Hodolic
- Department of Nuclear Medicine, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic.
| | - K Kairemo
- Department of Theragnostics, Docrates Cancer Center, Helsinki, Finland; Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Lee DY, Kim YI. Efficiency and Safety of Targeted Alpha Therapy in Metastatic Neuroendocrine Tumors: A Meta-analysis. Clin Nucl Med 2025; 50:e1-e6. [PMID: 39169519 DOI: 10.1097/rlu.0000000000005404] [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: 08/23/2024]
Abstract
PURPOSE Despite the effectiveness of 177 Lu-based peptide receptor radionuclide therapy in treating metastatic neuroendocrine tumors (NETs), disease progression posttreatment remains a significant challenge. Targeted alpha therapy (TAT) has emerged as a promising option for patients experiencing such progression. This study aims to assess the therapeutic efficiency and toxicity of TAT in patients with metastatic NET through a meta-analysis. PATIENTS AND METHODS We conducted a comprehensive search of PubMed, Embase, Cochrane Library, and CINAHL using relevant keywords. The analysis focused on the pooled proportions of objective response rate (ORR) and disease control rate (DCR) to determine therapeutic efficiency. We also evaluated the incidence of serious hematologic and renal adverse events (grade 3 or 4) to assess toxicity. A subgroup analysis was performed to identify factors influencing therapeutic outcomes. RESULTS Our meta-analysis included 7 studies comprising 162 patients. The results showed that TAT achieved ORR of 49.5% (95% confidence interval [CI]: 41.7%-57.4%) and DCR of 87.0% (95% CI: 72.1%-96.8%). The incidences of hematologic and renal toxicities were low, at 2.1% (95% CI: 0.5%-5.5%) and 3.4% (95% CI: 1.2%-7.3%), respectively. Subgroup analysis indicated consistent therapeutic efficiency across different variables, including prior 177 Lu-based peptide receptor radionuclide therapy treatment, 225 Ac-based TAT, absence of radiosensitizer, and methods of response evaluation, with ORR ranging from 46.6% to 57.1% and DCR from 82.0% to 91.5%. CONCLUSIONS TAT is an effective treatment for metastatic NET, demonstrating substantial disease control and response rates with minimal toxicity. These findings suggest that TAT is a viable therapeutic alternative for patients with metastatic NET.
Collapse
Affiliation(s)
- Dong Yun Lee
- From the Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
4
|
Gadens Zamboni C, Dundar A, Jain S, Kruzer M, Loeffler BT, Graves SA, Pollard JH, Mott SL, Dillon JS, Graham MM, Menda Y, Shariftabrizi A. Inter- and intra-tumoral heterogeneity on [ 68Ga]Ga-DOTA-TATE/[ 68Ga]Ga-DOTA-TOC PET/CT predicts response to [ 177Lu]Lu-DOTA-TATE PRRT in neuroendocrine tumor patients. EJNMMI REPORTS 2024; 8:39. [PMID: 39613925 DOI: 10.1186/s41824-024-00227-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/18/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Indices of tumor heterogeneity on somatostatin receptor PET/CT scans may potentially serve as predictive biomarkers of treatment efficacy in neuroendocrine tumor (NET) patients undergoing [177Lu]Lu-DOTA-TATE PRRT. METHODS NET patients who underwent [177Lu]Lu-DOTA-TATE therapy at the University of Iowa from August 2018 to February 2021 were retrospectively evaluated. Radiomic features on the pre-PRRT somatostatin receptor PET/CT were evaluated using a custom MIM Software® LesionID workflow. Conventional PET/CT metrics of tumor burden, such as somatostatin receptor expression and tumor volume, were calculated in addition to the indices of tumor heterogeneity for each lesion (intra-lesional) and then summarized across all lesions throughout the body (inter-lesional). Endpoints included post-PRRT 24-month time to progression (TTP) and overall survival (OS). Cox regression models were used to assess the predictive ability of the imaging factors on post-PRRT 24-month TTP and OS. LASSO-penalized Cox regression was used to build a multivariable model for each outcome. RESULTS Eighty patients with a mean age of 65.1 years were included, with most (71.3%) completing 4 cycles of PRRT. Median TTP was 19.1 months, and OS at 60 months was 50%. A large degree of variability between patients was evidenced for imaging features related to somatostatin receptor expression. On multivariable analysis, total receptor expression and mean liver-corrected SUVmean were selected for 24-month TTP. The model was not able to significantly predict progression (C-statistic = 0.58, 95% CI 0.50-0.62). Total receptor expression and mean skewness were selected for OS. The resulting model was able to significantly predict death (C-statistic = 0.62, 95% CI 0.53-0.67), but the predictive ability was limited, as evidenced by the low C-statistic. CONCLUSIONS Our exploratory analysis provides preliminary results showing that imaging indices of inter- and intra-tumor heterogeneity from pretreatment PET/CT images may potentially predict treatment efficacy in NET patients undergoing [177Lu]Lu-DOTA-TATE therapy. However, prospective evaluation in a larger cohort is needed to further assess whether a comprehensive characterization of tumor heterogeneity within a patient can help guide treatment decisions.
Collapse
Affiliation(s)
- Camila Gadens Zamboni
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Ayca Dundar
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Sanchay Jain
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | | | - Bradley T Loeffler
- Biostatistics Core, Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Stephen A Graves
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Janet H Pollard
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Sarah L Mott
- Biostatistics Core, Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Joseph S Dillon
- Department of Medicine-Hematology/Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Michael M Graham
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Yusuf Menda
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Ahmad Shariftabrizi
- Division of Nuclear Medicine, Department of Radiology-Room 3820-AJPP, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| |
Collapse
|
5
|
Abdrakhmanova D, Niemczyk K, Mukhamadieva G, Mustafin A, Bartosevic R. Tactics of surgical treatment of recurrent glomus tumors of the skull base: A clinical case. J Tissue Viability 2024; 33:895-902. [PMID: 39313404 DOI: 10.1016/j.jtv.2024.09.002] [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: 07/11/2023] [Revised: 08/26/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
AIM The purpose of the study is to build effective tactics of surgical treatment of pathology, and to increase the effectiveness of surgical treatment of patients with this diagnosis by optimising preoperative preparation, and the correct sequence of actions that will help prevent relapse. METHODS The leading approach to the study is the description of a clinical case, which will help to carefully consider this issue from all angles, analyse the methodology of patient examination, and build highly effective tactics of surgical intervention to eliminate tumour-like masses, minimising the risks of recurrence. The second clinical case was also considered, and a comparative analysis was carried out on certain parameters. RESULTS The paper presents a clinical case with a practical aspect of surgical treatment of the pathology. The issue of detailed symptoms and manifestations of the disease is disclosed, and the course of surgical interventions is described in stages, in which no nerve is damaged, no complications are caused, and the desired result is achieved. The peculiarity of this case is the recurrence of the disease, and the ineffectiveness of previous treatment methods, including embolisation of the neoplasm and antromastoidotomy. CONCLUSIONS The results obtained in the course of this study and the formulated conclusions are of great importance for surgeons who are faced with patients with the stated diagnosis.
Collapse
Affiliation(s)
- Dina Abdrakhmanova
- Department of Otolaryngology, Astana Medical University, 010000, 49A Beybitshilik Str., Astana, Kazakhstan.
| | - Kazimierz Niemczyk
- Department of Otolaryngology, Medical University of Warsaw, 02-091, 61 Zwirki i Wigury Str., Warsaw, Poland
| | - Gulmira Mukhamadieva
- Department of Otolaryngology, Astana Medical University, 010000, 49A Beybitshilik Str., Astana, Kazakhstan
| | - Adil Mustafin
- Department of Otolaryngology, Astana Medical University, 010000, 49A Beybitshilik Str., Astana, Kazakhstan
| | - Robert Bartosevic
- Department of Otolaryngology, Medical University of Warsaw, 02-091, 61 Zwirki i Wigury Str., Warsaw, Poland
| |
Collapse
|
6
|
Ria T, Roy R, Mandal US, Sk UH. Prospects of nano-theranostic approaches against breast and cervical cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189227. [PMID: 39612962 DOI: 10.1016/j.bbcan.2024.189227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
The bottleneck on therapeutics and diagnostics is removed by an alternate approach known as theranostics which combines both therapeutics and diagnostics within a single platform. Due to this "all in one" nature of theranostics, it is now extensively applied in the medicinal field mainly in cancer treatment over the conventional therapy. Recently, FDA approval of lutetium 177 (177Lu) DOTATATE and 177Lu-PSMA-based radionuclide theranostics are clinically used and very few theranostics specific to breast cancer are in clinical trials. In this review, we are willing to draw special attention to the application of theranostics in the most relevant cancers in women, the breast and the cervical as these cancers affect women harshly but talked very silently due to the social restrictions and discriminations mainly in rural areas of developing and under developing countries. This approach not only combines therapeutics and diagnostics but targeting moieties can also be accommodated for the precise medication. Herein, our main objective is to enlighten the broader aspects of different kinds of theranostic devices based on radioisotopes, nanoparticles, graphene quantum dots, dendrimers and their fruitful application against breast and cervical cancer. The development of synthetic nano-theranostics was reported by accommodating therapeutic drugs, imaging probes and targeting ligands through conjugation or encapsulation. The imaging modalities like optical fluorescence, photosensitizers and radiotracers are used to get the diagnostic images through NIR, PET, MRI and CT/SPECT to detect the progress of cancer non-invasively and also at the same time targeting ligands such as antibodies, proteins and peptides in attachment with the theranostics enhances the therapeutic efficacy in addition to the clarity in diagnostics. The applications of theranostics from the last decade with their present scenario in clinics and future perspectives, as well as the pitfalls with the hurdles that still leave questions to rethink from the root are also been discussed in this review.
Collapse
Affiliation(s)
- Tasnim Ria
- Department of Clinical and Translational Research, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700 026, India
| | - Rubi Roy
- Department of Clinical and Translational Research, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700 026, India
| | - Uma Sankar Mandal
- Department of Chemistry, Jadavpur University, Kolkata 700 032, India
| | - Ugir Hossain Sk
- Department of Clinical and Translational Research, Chittaranjan National Cancer Institute, 37 S.P. Mukherjee Road, Kolkata 700 026, India.
| |
Collapse
|
7
|
Piscopo L, Zampella E, Volpe F, Gaudieri V, Nappi C, Di Donna E, Clemente S, Varallo A, Scaglione M, Cuocolo A, Klain M. The Safety and Efficacy of Peptide Receptor Radionuclide Therapy for Gastro-Entero-Pancreatic Neuroendocrine Tumors: A Single Center Experience. Curr Oncol 2024; 31:5617-5629. [PMID: 39330044 PMCID: PMC11431754 DOI: 10.3390/curroncol31090416] [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: 07/23/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of the present study was to evaluate the safety and efficacy of radionuclide therapy with [177Lu]Lu-DOTA-TATE according to our single center experience at the University of Naples Federico II. For the present analysis, we considered 21 patients with progressive, advanced, well-differentiated G1 and G2 in patients with gastro-entero-pancreatic (GEP) neuroendocrine tumors (NETs) treated with [177Lu]Lu-DOTA-TATE according to the decisions of a multidisciplinary team. All patients underwent four cycles of 7-8 GBq of [177Lu]Lu-DOTA-TATE every 8 weeks. A whole-body scan (WBS) was performed 4, 48, and 168 h after each treatment. The dosimetry towards the organ at risk and target lesions was calculated. For each patient, renal and bone marrow parameters were evaluated before, during, and 3 months after the end of the treatment. Follow-up data were obtained and RECIST criteria were considered as the endpoint. Among 21 patients enrolled (mean age 65 ± 9 years); 17 (81%) were men and the small intestine was the most frequent location of disease (n = 12). A mild albeit significant variation (p < 0.05) in both platelets and white blood cell counts among all time points was observed, despite it disappearing 3 months after the end of the therapy. According to the RECIST criteria, 11 (55%) patients had a partial response to therapy and 8 (40%) had stable disease. Only one (5%) patient had disease progression 4 months after treatment. Our data confirm that [177Lu]Lu-DOTA is safe and effective in controlling the burden disease of G1/G2 GEP-NETs patients.
Collapse
Affiliation(s)
- Leandra Piscopo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Erica Di Donna
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Stefania Clemente
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Antonio Varallo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Mariano Scaglione
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy; (E.Z.); (F.V.); (V.G.); (C.N.); (E.D.D.); (S.C.); (A.V.); (A.C.); (M.K.)
| |
Collapse
|
8
|
Sozio SJ, Raynor W, Becker MC, Yudd A, Kempf JS. Carcinoid crisis in Lutetium-177-Dotatate therapy of neuroendocrine tumors: an overview of pathophysiology, risk factors, recognition, and treatment. EJNMMI REPORTS 2024; 8:29. [PMID: 39266864 PMCID: PMC11393224 DOI: 10.1186/s41824-024-00216-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/01/2024] [Indexed: 09/14/2024]
Abstract
PURPOSE Lutetium-177-Dotatate (Lutathera®) is a combined radionuclide-peptide that is FDA-approved for the treatment of well-differentiated, somatostatin receptor-positive, gastroenteropancreatic neuroendocrine tumors. Carcinoid crisis is a rare, but potentially life-threatening risk of this radiopharmaceutical, of which prompt recognition and treatment is essential to reducing morbidity. This manuscript provides an overview of the topic to promote awareness of this adverse event, with emphasis on early recognition and management. In addition, we present our institution's experience with Lutetium-177-Dotatate-associated complications across a five-year period. METHODS A literature review of lutetium-177-dotatate therapy and its potential implication of carcinoid crisis was performed. Additionally, a review of our institution's experience is presented. RESULTS The incidence of carcinoid crisis induced by Lutetium-177-Dotatate therapy is estimated to range between 1 and 2% of treatment recipients. Those who have tumors located within the midgut, higher tumor burden, and the presence of metastasis have an increased risk of developing carcinoid crisis, among other risk factors. Carcinoid crisis is most often encountered within 12-48 h of receiving the first treatment dose, with the most common symptoms being nausea/vomiting, flushing, and diarrhea. CONCLUSION Carcinoid crisis is a rare but potentially life-threatening complication of Lutetium-177-Dotatate therapy. Knowledge of risk factors and prompt recognition of symptoms is essential to successful treatment, with early initiation of intravenous octreotide serving a critical step in reducing morbidity of this adverse event.
Collapse
Affiliation(s)
- Stephen J Sozio
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - William Raynor
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Murray C Becker
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Anthony Yudd
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jeffrey S Kempf
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
9
|
Wild D, Grønbæk H, Navalkissoor S, Haug A, Nicolas GP, Pais B, Ansquer C, Beauregard JM, McEwan A, Lassmann M, Pennestri D, Volteau M, Lenzo NP, Hicks RJ. A phase I/II study of the safety and efficacy of [ 177Lu]Lu-satoreotide tetraxetan in advanced somatostatin receptor-positive neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2023; 51:183-195. [PMID: 37721581 PMCID: PMC10684626 DOI: 10.1007/s00259-023-06383-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/02/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE We present the results of an open-label, phase I/II study evaluating the safety and efficacy of the novel somatostatin receptor (SSTR) antagonist [177Lu]Lu-satoreotide tetraxetan in 40 patients with previously treated, progressive neuroendocrine tumours (NETs), in which dosimetry was used to guide maximum administered activity. METHODS This study was conducted in two parts. Part A consisted of 15 patients who completed three cycles of [177Lu]Lu-satoreotide tetraxetan at a fixed administered activity and peptide amount per cycle (4.5 GBq/300 µg). Part B, which included 25 patients who received one to five cycles of [177Lu]Lu-satoreotide tetraxetan, evaluated different administered activities (4.5 or 6.0 GBq/cycle) and peptide amounts (300, 700, or 1300 μg/cycle), limited to a cumulative absorbed radiation dose of 23 Gy to the kidneys and 1.5 Gy to the bone marrow. RESULTS Median cumulative administered activity of [177Lu]Lu-satoreotide tetraxetan was 13.0 GBq over three cycles (13.1 GBq in part A and 12.9 GBq in part B). Overall, 17 (42.5%) patients experienced grade ≥ 3 treatment‑related adverse events; the most common were lymphopenia, thrombocytopenia, and neutropenia. No grade 3/4 nephrotoxicity was observed. Two patients developed myeloid neoplasms considered treatment related by the investigator. Disease control rate for part A and part B was 94.7% (95% confidence interval [CI]: 82.3-99.4), and overall response rate was 21.1% (95% CI: 9.6-37.3). CONCLUSION [177Lu]Lu-satoreotide tetraxetan, administered at a median cumulative activity of 13.0 GBq over three cycles, has an acceptable safety profile with a promising clinical response in patients with progressive, SSTR-positive NETs. A 5-year long-term follow-up study is ongoing. TRIAL REGISTRATION ClinicalTrials.gov, NCT02592707. Registered October 30, 2015.
Collapse
Affiliation(s)
- Damian Wild
- Division of Nuclear Medicine, ENETS Centre of Excellence, University Hospital Basel, Basel, Switzerland.
| | - Henning Grønbæk
- Department of Hepatology & Gastroenterology, ENETS Centre of Excellence, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Shaunak Navalkissoor
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free London NHS Foundation Trust, London, UK
| | - Alexander Haug
- Department of Radiology and Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Guillaume P Nicolas
- Division of Nuclear Medicine, ENETS Centre of Excellence, University Hospital Basel, Basel, Switzerland
| | - Ben Pais
- SRT-Biomedical B.V., Soest, Netherlands
- Ariceum Therapeutics GmbH, Berlin, Germany
| | | | | | | | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Nat P Lenzo
- GenesisCare, East Fremantle, Australia
- Department of Medicine, Curtin University, Perth, Australia
| | - Rodney J Hicks
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Melbourne, Australia
- Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Australia
| |
Collapse
|
10
|
Halfdanarson TR, Mallak N, Paulson S, Chandrasekharan C, Natwa M, Kendi AT, Kennecke HF. Monitoring and Surveillance of Patients with Gastroenteropancreatic Neuroendocrine Tumors Undergoing Radioligand Therapy. Cancers (Basel) 2023; 15:4836. [PMID: 37835530 PMCID: PMC10571645 DOI: 10.3390/cancers15194836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Radioligand therapy (RLT) with [177Lu]Lu-DOTA-TATE is a standard of care for adult patients with somatostatin-receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Taking advantage of this precision nuclear medicine approach requires diligent monitoring and surveillance, from the use of diagnostic SSTR-targeted radioligand imaging for the selection of patients through treatment and assessments of response. Published evidence-based guidelines assist the multidisciplinary healthcare team by providing acceptable approaches to care; however, the sheer heterogeneity of GEP-NETs can make these frameworks difficult to apply in individual clinical circumstances. There are also contradictions in the literature regarding the utility of novel approaches in monitoring and surveilling patients with GEP-NETs receiving RLT. This article discusses the emerging evidence on imaging, clinical biochemistry, and tumor assessment criteria in the management of patients receiving RLT for GEP-NETs; additionally, it documents our own best practices. This allows us to offer practical guidance on how to effectively implement monitoring and surveillance measures to aid patient-tailored clinical decision-making.
Collapse
Affiliation(s)
| | - Nadine Mallak
- Division of Molecular Imaging and Therapy, Oregon Health and Science University, Portland, OR 97239, USA;
| | | | | | - Mona Natwa
- Langone Health, New York University, New York, NY 10016, USA
| | | | | |
Collapse
|
11
|
Corlett A, Pinson JA, Rahimi MN, Zuylekom JV, Cullinane C, Blyth B, Thompson PE, Hutton CA, Roselt PD, Haskali MB. Development of Highly Potent Clinical Candidates for Theranostic Applications against Cholecystokinin-2 Receptor Positive Cancers. J Med Chem 2023; 66:10289-10303. [PMID: 37493526 DOI: 10.1021/acs.jmedchem.3c00377] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Peptide receptor radionuclide therapy (PRRT) is a promising form of systemic radiation therapy designed to eradicate cancer. Cholecystokinin-2 receptor (CCK2R) is an important molecular target that is highly expressed in a range of cancers. This study describes the synthesis and in vivo characterization of a novel series of 177Lu-labeled peptides ([177Lu]Lu-2b-4b) in comparison with the reference CCK2R-targeting peptide CP04 ([177Lu]Lu-1b). [177Lu]Lu-1b-4b showed high chemical purity (HPLC ≥ 94%), low Log D7.4 (-4.09 to -4.55) with strong binding affinity to CCK2R (KD 0.097-1.61 nM), and relatively high protein binding (55.6-80.2%) and internalization (40-67%). Biodistribution studies of the novel 177Lu-labeled peptides in tumors (AR42J and A431-CCK2R) showed uptake one- to eight-fold greater than the reference compound CP04 at 1, 24, and 48 h. Rapid clearance and high tumor uptake and retention were established for [177Lu]Lu-2b-4b, making these compounds excellent candidates for theranostic applications against CCK2R-expressing tumors.
Collapse
Affiliation(s)
- Alicia Corlett
- Department of Nuclear Medicine, The Royal Melbourne Hospital, Parkville, Victoria, 3000, Australia
| | - Jo-Anne Pinson
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marwa N Rahimi
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jessica Van Zuylekom
- Models of Cancer Translational Research Centre, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Carleen Cullinane
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
- The Centre for Molecular Imaging and Translational Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Benjamin Blyth
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
- Models of Cancer Translational Research Centre, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Philip E Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University (Parkville Campus), Parkville, Victoria 3052, Australia
| | - Craig A Hutton
- School of Chemistry, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Peter D Roselt
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Mohammad B Haskali
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
12
|
Parghane RV, Basu S. PSMA-targeted radioligand therapy in prostate cancer: current status and future prospects. Expert Rev Anticancer Ther 2023; 23:959-975. [PMID: 37565281 DOI: 10.1080/14737140.2023.2247562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/19/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION The prostate-specific membrane antigen (PSMA) targeted radioligand therapy (PRLT) for the treatment of metastatic castration-resistant prostate cancer (mCRPC) patients has generated significant interest among the oncologic community, with several publications documenting good response rates and survival benefits with low toxicity profiles. AREAS COVERED Indications, patient preparation, dose administration, post-treatment imaging, dosimetry, and side effect profiles of 177Lu-PSMA-617 are discussed in this article. We also discuss results from prospective studies, major retrospective studies, meta-analyses, clinical trials, and mentioned major ongoing clinical trials on PRLT. We have also portrayed our own experiences and future perspectives on PRLT. EXPERT OPINION For PRLT, PSMA-617 and PSMA-I&T molecules have revolutionized the theranostic approach in the management of advanced prostate cancer, with solid backing from several published articles showing favorable outcomes and an excellent safety profile of 177Lu-PSMA-617. Improvement in quality of life and survival was seen in the majority of mCRPC patients after 177Lu-PSMA-617 PRLT. Patients with good performance status, asymptomatic, only lymph node metastases, high PSMA expressing lesions, and no discordant FDG avid lesions have a longer survival after 177Lu-PSMA-617 PRLT than patients with poor performance status, symptomatic, hepatic, brain, and skeletal metastases, discordant PSMA, and FDG-avid lesions. Docetaxel and cabazitaxel are approved treatments for mCRPC patients. 177Lu-PSMA-617 is approved as a third-line systemic treatment for mCRPC patients with failure to respond to androgen receptor pathway inhibitors and docetaxel therapy. PRLT is a safe and effective alternative to cabazitaxel (third-line systemic treatment), but it has a higher cost. 177Lu-PSMA-617 could be a more efficient therapeutic option for mCRPC patients as first-line or combined therapy, and it may be a useful therapeutic option for the treatment of metastatic hormone-sensitive prostate cancer (mHSPC) patients. Several clinical studies and clinical trials on PRLT are currently underway. In the future, the results of these trials will be helpful in evolving treatment strategies for prostate cancer patients.
Collapse
Affiliation(s)
- Rahul V Parghane
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Mumbai, India
- Radiation Medicine Centre (BARC), Homi Bhabha National Institute, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Mumbai, India
- Radiation Medicine Centre (BARC), Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
13
|
Burkett BJ, Bartlett DJ, McGarrah PW, Lewis AR, Johnson DR, Berberoğlu K, Pandey MK, Packard AT, Halfdanarson TR, Hruska CB, Johnson GB, Kendi AT. A Review of Theranostics: Perspectives on Emerging Approaches and Clinical Advancements. Radiol Imaging Cancer 2023; 5:e220157. [PMID: 37477566 PMCID: PMC10413300 DOI: 10.1148/rycan.220157] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 07/22/2023]
Abstract
Theranostics is the combination of two approaches-diagnostics and therapeutics-applied for decades in cancer imaging using radiopharmaceuticals or paired radiopharmaceuticals to image and selectively treat various cancers. The clinical use of theranostics has increased in recent years, with U.S. Food and Drug Administration (FDA) approval of lutetium 177 (177Lu) tetraazacyclododecane tetraacetic acid octreotate (DOTATATE) and 177Lu-prostate-specific membrane antigen vector-based radionuclide therapies. The field of theranostics has imminent potential for emerging clinical applications. This article reviews critical areas of active clinical advancement in theranostics, including forthcoming clinical trials advancing FDA-approved and emerging radiopharmaceuticals, approaches to dosimetry calculations, imaging of different radionuclide therapies, expanded indications for currently used theranostic agents to treat a broader array of cancers, and emerging ideas in the field. Keywords: Molecular Imaging, Molecular Imaging-Cancer, Molecular Imaging-Clinical Translation, Molecular Imaging-Target Development, PET/CT, SPECT/CT, Radionuclide Therapy, Dosimetry, Oncology, Radiobiology © RSNA, 2023.
Collapse
Affiliation(s)
- Brian J. Burkett
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - David J. Bartlett
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Patrick W. McGarrah
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Akeem R. Lewis
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Derek R. Johnson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Kezban Berberoğlu
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Mukesh K. Pandey
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Annie T. Packard
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Thorvardur R. Halfdanarson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Carrie B. Hruska
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - Geoffrey B. Johnson
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| | - A. Tuba Kendi
- From the Department of Radiology (B.J.B., D.J.B., D.R.J., M.K.P.,
A.T.P., C.B.H., G.B.J., A.T.K.) and Division of Medical Oncology (P.W.M.,
A.R.L., T.R.H.), Mayo Clinic, 200 First St SW, Rochester, MN 55905; and
Department of Nuclear Medicine, Anadolu Medical Center, Gebze/Kocaeli, Turkey
(K.B.)
| |
Collapse
|
14
|
Carr HS, Zuo Y, Frost JA. The Wnt pathway protein Dvl1 targets somatostatin receptor 2 for lysosome-dependent degradation. J Biol Chem 2023; 299:104645. [PMID: 36965619 PMCID: PMC10164914 DOI: 10.1016/j.jbc.2023.104645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/27/2023] Open
Abstract
The Somatostatin receptor 2 (Sstr2) is a heterotrimeric G protein-coupled receptor that is highly expressed in neuroendocrine tumors and is a common pharmacological target for intervention. Unfortunately, not all neuroendocrine tumors express Sstr2, and Sstr2 expression can be downregulated with prolonged agonist use. Sstr2 is rapidly internalized following agonist stimulation and, in the short term, is quantitatively recycled back to the plasma membrane. However, mechanisms controlling steady state expression of Sstr2 in the absence of agonist are less well described. Here, we show that Sstr2 interacts with the Wnt pathway protein Dvl1 in a ligand-independent manner to target Sstr2 for lysosomal degradation. Interaction of Sstr2 with Dvl1 does not affect receptor internalization, recycling, or signaling to adenylyl cyclase but does suppress agonist-stimulated ERK1/2 activation. Importantly, Dvl1-dependent degradation of Sstr2 can be stimulated by overexpression of Wnts and treatment of cells with Wnt pathway inhibitors can boost Sstr2 expression in neuroendocrine tumor cells. Taken together, this study identifies for the first time a mechanism that targets Sstr2 for lysosomal degradation that is independent of Sstr2 agonist and can be potentiated by Wnt ligand. Intervention in this signaling mechanism has the potential to elevate Sstr2 expression in neuroendocrine tumors and enhance Sstr2-directed therapies.
Collapse
Affiliation(s)
- Heather S Carr
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA
| | - Yan Zuo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA
| | - Jeffrey A Frost
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA.
| |
Collapse
|
15
|
Utility of PET Scans in the Diagnosis and Management of Gastrointestinal Tumors. Dig Dis Sci 2022; 67:4633-4653. [PMID: 35908126 DOI: 10.1007/s10620-022-07616-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
|
16
|
Zamponi V, La Salvia A, Tarsitano MG, Mikovic N, Rinzivillo M, Panzuto F, Giannetta E, Faggiano A, Mazzilli R. Effect of Neuroendocrine Neoplasm Treatment on Human Reproductive Health and Sexual Function. J Clin Med 2022; 11:jcm11143983. [PMID: 35887747 PMCID: PMC9324753 DOI: 10.3390/jcm11143983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/02/2023] Open
Abstract
Neuroendocrine neoplasms (NEN) are characterized by a wide clinical heterogeneity and biological variability, with slow progression and long survival in most cases. Although these tumors can affect young adults, there are few studies that focus on the sexual and reproductive system. The aim of this review was to evaluate the effect of NEN treatment, including somatostatin analogues (SSA), targeted therapy (Everolimus and Sunitinib), radiolabeled-SSA and chemotherapy, on male and female reproductive systems and sexual function. This narrative review was performed for all available prospective and retrospective studies, case reports and review articles published up to March 2022 in PubMed. To date, few data are available on the impact of SSA on human fertility and most of studies come from acromegalic patients. However, SSAs seem to cross the blood–placental barrier; therefore, pregnancy planning is strongly recommended. Furthermore, the effect of targeted therapy on reproductive function is still undefined. Conversely, chemotherapy has a well-known negative impact on male and female fertility. The effect of temozolomide on reproductive function is still undefined, even if changes in semen parameters after the treatment have been described. Finally, very few data are available on the sexual function of NEN treatment.
Collapse
Affiliation(s)
- Virginia Zamponi
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Anna La Salvia
- Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: ; Tel.: +39-0652665698
| | - Maria Grazia Tarsitano
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Nevena Mikovic
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Maria Rinzivillo
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
| | - Francesco Panzuto
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| |
Collapse
|
17
|
Ramonaheng K, van Staden JA, du Raan H. Accuracy of two dosimetry software programs for 177Lu radiopharmaceutical therapy using voxel-based patient-specific phantoms. Heliyon 2022; 8:e09830. [PMID: 35865988 PMCID: PMC9293745 DOI: 10.1016/j.heliyon.2022.e09830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/18/2022] [Accepted: 06/24/2022] [Indexed: 12/01/2022] Open
Abstract
Purpose Virtual dosimetry using voxel-based patient-specific phantoms and Monte Carlo (MC) simulations offer the advantage of having a gold standard against which absorbed doses may be benchmarked to establish the dosimetry accuracy. Furthermore, these reference values assist in investigating the accuracy of the absorbed dose methodologies from different software programs. Therefore, this study aimed to compare the accuracy of the absorbed doses computed using LundADose and OLINDA/EXM 1.0. Methods The accuracy was based on 177Lu-DOTATATE distributions of three voxel-based phantoms. SPECT projection images were simulated for 1, 24, 96, and 168 h post-administration and reconstructed with LundADose using 3D OS-EM reconstruction. Mono-exponential curves were fitted to the bio-kinetic data for the kidneys, liver, spleen, and tumours resulting in SPECT time-integrated activity (SPECT-TIA). The SPECT-TIA were used to compute mean absorbed doses using LundADose (LND-DSPECT) and OLINDA (OLINDA-DSPECT) for the organs. Pre-defined true activity images, were used to obtain TRUE-TIA and, together with full MC simulations, computed the true doses (MC-DTrue). The dosimetry accuracy was assessed by comparing LND-DSPECT and OLINDA-DSPECT to MC-DTrue. Results Overall, the results presented an overestimation of the mean absorbed dose by LND-DSPECT compared to the MC-DTrue with a dosimetry accuracy ≤6.6%. This was attributed to spill-out activity from the reconstructed LND-DSPECT, resulting in a higher dose contribution than the MC-DTrue. There was a general underestimation (<8.1%) of OLINDA-DSPECT compared to MC-DTrue attributed to the geometrical difference in shape between the voxel-based phantoms and the OLINDA models. Furthermore, OLINDA-DSPECT considers self-doses while MC-DTrue reflects self-doses plus cross-doses. Conclusion The better than 10% accuracy suggests that the mean dose values obtained with LND-DSPECT and OLINDA-DSPECT approximate the true values. The mean absorbed doses of the two software programs, and the gold standard were comparable. This work shall be of use for optimising 177Lu dosimetry for clinical applications.
Collapse
Affiliation(s)
- Keamogetswe Ramonaheng
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, P.O. Box 339, Bloemfontein, 9300, South Africa
| | - Johannes A van Staden
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, P.O. Box 339, Bloemfontein, 9300, South Africa
| | - Hanlie du Raan
- Department of Medical Physics, Faculty of Health Sciences, University of the Free State, P.O. Box 339, Bloemfontein, 9300, South Africa
| |
Collapse
|
18
|
Simón M, Jørgensen JT, Khare HA, Christensen C, Nielsen CH, Kjaer A. Combination of [ 177Lu]Lu-DOTA-TATE Targeted Radionuclide Therapy and Photothermal Therapy as a Promising Approach for Cancer Treatment: In Vivo Studies in a Human Xenograft Mouse Model. Pharmaceutics 2022; 14:pharmaceutics14061284. [PMID: 35745856 PMCID: PMC9227845 DOI: 10.3390/pharmaceutics14061284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) relies on α- and β-emitting radionuclides bound to a peptide that commonly targets somatostatin receptors (SSTRs) for the localized killing of tumors through ionizing radiation. A Lutetium-177 (177Lu)-based probe linked to the somatostatin analog octreotate ([177Lu]Lu-DOTA-TATE) is approved for the treatment of certain SSTR-expressing tumors and has been shown to improve survival. However, a limiting factor of PRRT is the potential toxicity derived from the high doses needed to kill the tumor. This could be circumvented by combining PRRT with other treatments for an enhanced anti-tumor effect. Photothermal therapy (PTT) relies on nanoparticle-induced hyperthermia for cancer treatment and could be a useful add-on to PRRT. Here, we investigate a strategy combining [177Lu]Lu-DOTA-TATE PRRT and nanoshell (NS)-based PTT for the treatment of SSTR-expressing small-cell lung tumors in mice. Our results showed that the combination treatment improved survival compared to PRRT alone, but only when PTT was performed one day after [177Lu]Lu-DOTA-TATE injection (one of the timepoints examined), showcasing the effect of treatment timing in relation to outcome. Furthermore, the combination treatment was well-tolerated in the mice. This indicates that strategies involving NS-based PTT as an add-on to PRRT could be promising and should be investigated further.
Collapse
Affiliation(s)
- Marina Simón
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
| | - Harshvardhan A. Khare
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
| | - Camilla Christensen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
- Minerva Imaging, 3650 Ølstykke, Denmark
| | - Carsten Haagen Nielsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
- Minerva Imaging, 3650 Ølstykke, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.S.); (J.T.J.); (H.A.K.); (C.C.); (C.H.N.)
- Correspondence:
| |
Collapse
|
19
|
Early Complications of Radioisotope Therapy with Lutetium-177 and Yttrium-90 in Patients with Neuroendocrine Neoplasms-A Preliminary Study. J Clin Med 2022; 11:jcm11040919. [PMID: 35207193 PMCID: PMC8874379 DOI: 10.3390/jcm11040919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) constitute a heterogenous group of tumors originating from neuroendocrine cells scattered throughout the body. Peptide Receptor Radionuclide Therapy (PRRT) is a treatment of choice of unresectable metastasized progressive and well-differentiated NENs. The aim of the study was to assess early bone marrow and kidney injury after administration of Lutetium-177 or Lutetium-177 combined with Yttrium-90. Thirty-one patients received treatment with [177Lu]Lu-DOTATATE with the activity of 7.4 GBq. Eleven patients received tandem treatment with [90Y]Y-DOTATATE with the activity of 1.85 GBq + [177Lu]Lu-DOTATATE with the activity of 1.85 GBq. After PRRT a significant decrease in leukocyte, neutrophil, and lymphocyte counts was noted. Tandem treatment demonstrated a more marked decrease in white blood cell count compared to Lutetium-177 therapy only. Conversely, no significant influence on glomerular filtration was found in this assessment. However, PRRT triggered acute renal tubule dysfunction, regardless of the treatment type. Regarding the acute complications, PRRT appeared to be a safe modality in the treatment of patients with NEN.
Collapse
|
20
|
Naik M, Al-Nahhas A, Khan SR. Treatment of Neuroendocrine Neoplasms with Radiolabeled Peptides-Where Are We Now. Cancers (Basel) 2022; 14:761. [PMID: 35159027 PMCID: PMC8833798 DOI: 10.3390/cancers14030761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been one of the most successful and exciting examples of theranostics in nuclear medicine in recent decades and is now firmly embedded in many treatment algorithms for unresectable or metastatic neuroendocrine neoplasms (NENs) worldwide. It is widely considered to be an effective treatment for well- or moderately differentiated neoplasms, which express high levels of somatostatin receptors that can be selectively targeted. This review article outlines the scientific basis of PRRT in treatment of NENs and describes its discovery dating back to the early 1990s. Early treatments utilizing Indium-111, a γ-emitter, showed promise in reduction in tumor size and improvement in biochemistry, but were also met with high radiation doses and myelotoxic and nephrotoxic effects. Subsequently, stable conjugation of DOTA-peptides with β-emitting radionuclides, such as Yttrium-90 and Lutetium-177, served as a breakthrough for PRRT and studies highlighted their potential in eliciting progression-free survival and quality of life benefits. This article will also elaborate on the key trials which paved the way for its approval and will discuss therapeutic considerations, such as patient selection and administration technique, to optimize its use.
Collapse
Affiliation(s)
- Mitesh Naik
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| | | | - Sairah R. Khan
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| |
Collapse
|
21
|
Griffiths GL, Vasquez C, Escorcia F, Clanton J, Lindenberg L, Mena E, Choyke PL. Translating a radiolabeled imaging agent to the clinic. Adv Drug Deliv Rev 2022; 181:114086. [PMID: 34942275 PMCID: PMC8889912 DOI: 10.1016/j.addr.2021.114086] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/30/2021] [Accepted: 12/16/2021] [Indexed: 02/03/2023]
Abstract
Molecular Imaging is entering the most fruitful, exciting period in its history with many new agents under development, and several reaching the clinic in recent years. While it is unusual for just one laboratory to take an agent from initial discovery through to full clinical approval the steps along the way are important to understand for all interested participants even if one is not involved in the entire process. Here, we provide an overview of these processes beginning at discovery and preclinical validation of a new molecular imaging agent and using as an exemplar a low molecular weight disease-specific targeted positron emission tomography (PET) agent. Compared to standard drug development requirements, molecular imaging agents may benefit from a regulatory standpoint from their low mass administered doses, they nonetheless still need to go through a series of well-defined steps before they can be considered for Phase 1 human testing. After outlining the discovery and preclinical validation approaches, we will also discuss the nuances of Phase 1, Phase 2 and Phase 3 studies that may culminate in an FDA general use approval. Finally, some post-approval aspects of novel molecular imaging agents are considered.
Collapse
Affiliation(s)
- Gary L. Griffiths
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD
| | - Crystal Vasquez
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Freddy Escorcia
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | | | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD
| |
Collapse
|
22
|
Miller C, Rousseau J, Ramogida CF, Celler A, Rahmim A, Uribe CF. Implications of physics, chemistry and biology for dosimetry calculations using theranostic pairs. Theranostics 2022; 12:232-259. [PMID: 34987643 PMCID: PMC8690938 DOI: 10.7150/thno.62851] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022] Open
Abstract
Theranostics is an emerging paradigm that combines imaging and therapy in order to personalize patient treatment. In nuclear medicine, this is achieved by using radiopharmaceuticals that target identical molecular targets for both imaging (using emitted gamma rays) and radiopharmaceutical therapy (using emitted beta, alpha or Auger-electron particles) for the treatment of various diseases, such as cancer. If the therapeutic radiopharmaceutical cannot be imaged quantitatively, a “theranostic pair” imaging surrogate can be used to predict the absorbed radiation doses from the therapeutic radiopharmaceutical. However, theranostic dosimetry assumes that the pharmacokinetics and biodistributions of both radiopharmaceuticals in the pair are identical or very similar, an assumption that still requires further validation for many theranostic pairs. In this review, we consider both same-element and different-element theranostic pairs and attempt to determine if factors exist which may cause inaccurate dose extrapolations in theranostic dosimetry, either intrinsic (e.g. chemical differences) or extrinsic (e.g. injecting different amounts of each radiopharmaceutical) to the radiopharmaceuticals. We discuss the basis behind theranostic dosimetry and present common theranostic pairs and their therapeutic applications in oncology. We investigate general factors that could create alterations in the behavior of the radiopharmaceuticals or the quantitative accuracy of imaging them. Finally, we attempt to determine if there is evidence showing some specific pairs as suitable for theranostic dosimetry. We show that there are a variety of intrinsic and extrinsic factors which can significantly alter the behavior among pairs of radiopharmaceuticals, even if they belong to the same chemical element. More research is needed to determine the impact of these factors on theranostic dosimetry estimates and on patient outcomes, and how to correctly account for them.
Collapse
|
23
|
Terapia con péptidos radiomarcados con [177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Prado-Wohlwend S, Bernal-Vergara JC, Utrera-Costero A, Cañón-Sánchez JR, Agudelo-Cifuentes M, Bello-Arques P. Peptide receptor radionuclide therapy with [ 177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2021; 41:55-65. [PMID: 34920969 DOI: 10.1016/j.remnie.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022]
Abstract
This continuing education aims to present in a clear and easy-to-understand way, the biology of neuroendocrine tumors (NETs), the characteristics of somatostatin receptors, the selection of patients for radiolabelled peptide therapy (PRRT), the inclusion criteria to benefit from treatment with the minimum possible adverse effects, the administration protocol, follow-up and response evaluation. The functional imaging studies necessary to explore the biology of the tumor and to individualize the treatment are also carried out, and constitute the cornerstone for the development of teragnosis. Clinical trials are being developed to better define the position of PRRT within the broad therapeutic options, and among the future perspectives, there are several lines of research to improve the objective response rate and survival with PRRT, focused on the development of new agonists and somatostatin receptor antagonists, new radionuclides and radiosensitizing combination therapies. In conclusion, PRRT is a great therapeutic, well-tolerated and safe tool with generally mild and self-limited acute side effects, that must be sequenced at the best moment of the evolution of the disease of patients with NET. Candidate patients for PRRT should always be evaluated by a multidisciplinary clinical committee.
Collapse
Affiliation(s)
- S Prado-Wohlwend
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain.
| | - J C Bernal-Vergara
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - A Utrera-Costero
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - J R Cañón-Sánchez
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M Agudelo-Cifuentes
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - P Bello-Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | -
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
25
|
Breton JM, Arkun K, Tischler AS, Qamar AS, Sillman JS, Heilman CB. Clinical and histopathological principles for the diagnosis of a recurrent paraganglioma of the jugular foramen initially diagnosed as a middle ear adenoma: illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2021; 2:CASE21307. [PMID: 36131578 PMCID: PMC9563646 DOI: 10.3171/case21307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Paragangliomas (PGLs) are rare neoplasms that may be associated with hereditary PGL syndromes and variable risk of metastasis. Middle ear adenomas are extremely rare tumors with no known hereditary predisposition and extremely low risk of metastasis. Although often easily differentiated, they may share clinical and pathological features that misdirect and confuse the diagnosis. OBSERVATIONS The authors discussed a 35-year-old woman with left-sided hearing loss and bleeding from the external ear canal who presented to an outside hospital. She underwent resection of a middle ear and mastoid mass, initially diagnosed as a middle ear adenoma with neuroendocrine features, with later mastoidectomy and ligation of the sigmoid sinus with microsurgical excision of persistent tumor in the jugular foramen and temporal bone. Histopathologically, her tumor was vascular, composed of benign-appearing epithelioid cells with “salt and pepper” neuroendocrine chromatin arranged in vague nests. Lesional cells were GATA3-immunopositive, glucagon-negative, and succinate dehydrogenase-immunonegative, consistent with PGL rather than middle ear adenoma, and required further workup for hereditary PGL syndromes. LESSONS This case demonstrates potential challenges in differentiating a PGL from a middle ear adenoma. The authors offer clinical, histopathological, and imaging principles to aid in diagnosis and workup.
Collapse
Affiliation(s)
- Jeffrey M. Breton
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
| | - Knarik Arkun
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
- Departments of Pathology and Laboratory Medicine and
| | | | | | | | - Carl B. Heilman
- Department of Neurosurgery, Tufts Medical Center and Tufts University School of Medicine, Boston, Massachusetts; and
| |
Collapse
|
26
|
Hosono M, Takenaka M, Monzen H, Tamura M, Kudo M, Nishimura Y. Cumulative radiation doses from recurrent PET/CT examinations. Br J Radiol 2021; 94:20210388. [PMID: 34111964 PMCID: PMC9328066 DOI: 10.1259/bjr.20210388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Positron emission tomography (PET–CT) is an essential imaging modality for the management of various diseases. Increasing numbers of PET–CT examinations are carried out across the world and deliver benefits to patients; however, there are concerns about the cumulative radiation doses from these examinations in patients. Compared to the radiation exposure delivered by CT, there have been few reports on the frequency of patients with a cumulative effective radiation dose of ≥100 mSv from repeated PET–CT examinations. The emerging dose tracking system facilitates surveys on patient cumulative doses by PET–CT because it can easily wrap up exposure doses of PET radiopharmaceuticals and CT. Regardless of the use of a dose tracking system, implementation of justification for PET–CT examinations and utilisation of dose reduction measures are key issues in coping with the cumulative dose in patients. Despite all the advantages of PET/MRI such as eliminating radiation exposure from CT and providing good tissue contrast in MRI, it is expensive and cannot be introduced at every facility; thus, it is still necessary to utilise PET–CT with radiation reduction measures in most clinical situations.
Collapse
Affiliation(s)
- Makoto Hosono
- Department of Radiation Oncology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology, Faculty of Medicine, Kindai University, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Hajime Monzen
- . Department of Medical Physics, Graduate School of Medical Sciences, Kindai University, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Mikoto Tamura
- . Department of Medical Physics, Graduate School of Medical Sciences, Kindai University, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology, Faculty of Medicine, Kindai University, Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| | - Yasumasa Nishimura
- Department of Radiation Oncology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
27
|
Neuroendocrine Tumor Theranostics: An Update and Emerging Applications in Clinical Practice. AJR Am J Roentgenol 2021; 217:495-506. [PMID: 34076455 DOI: 10.2214/ajr.20.23349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE. Theranostics have shown great promise for delivering precision medicine, particularly in neuroendocrine tumors (NETs). The clinical applications of radiolabeled somatostatin analogues in imaging and radionuclide therapy have been rapidly increasing over the past 2 decades and are currently integrated into the management guidelines of NETs. This article summarizes the available literature on different somatostatin receptor-targeting radiopharmaceuticals with theranostic potential in NETs, pheochromocytomas, and paragangliomas. We discuss the clinical application, administration, and toxicity of recent FDA-approved radionuclide therapies, including 177Lu-DOTATATE in advanced gastroenteropancreatic NETs and 131I-MIBG in advanced paragangliomas and pheochromocytomas. CONCLUSION. Several studies support the safety and clinical efficacy of peptide receptor radionuclide therapies in disease control and quality-of-life improvement in patients with NETs and report potential benefits of combined radionuclide treatment approaches. The utility and pitfalls of functional imaging in therapy response assessment and surveillance of NETs remain to be established.
Collapse
|
28
|
Sanli Y, Simsek DH, Sanli O, Subramaniam RM, Kendi AT. 177Lu-PSMA Therapy in Metastatic Castration-Resistant Prostate Cancer. Biomedicines 2021; 9:430. [PMID: 33921146 PMCID: PMC8071500 DOI: 10.3390/biomedicines9040430] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
The aim of this narrative review is to evaluate the current status of 177Lu-PSMA (prostate specific membrane antigen) therapy for metastatic castration-resistant prostate cancer (mCRPC) in the light of the current literature. We also addressed patient preparation, therapy administration and side effect profiles. 177Lu-PSMA therapy efficacy was assessed by using prospective trials, meta-analyses and major retrospective trials. Predictors of efficacy were also mentioned. Although there are some different approaches regarding the use of 177Lu-PSMA therapy in different countries, this type of therapy is generally safe, with a low toxicity profile. From the oncological point of view, a PSA (prostate specific antigen) decline of ≥50% was seen in 10.6-69% of patients with mCRPC; whereas progression-free survival (PFS) was reported to be 3-13.7 months in different studies. Consequently, 177Lu-PSMA therapy is a promising treatment in patients with mCRPC, with good clinical efficacy, even in heavily pretreated patients with multiple lines of systemic therapy. Currently, there are ongoing clinical trials in the United States, including a phase III multicenter FDA registration trial.
Collapse
Affiliation(s)
- Yasemin Sanli
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34093, Turkey; (Y.S.); (D.H.S.)
| | - Duygu Has Simsek
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34093, Turkey; (Y.S.); (D.H.S.)
| | - Oner Sanli
- Department of Urology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34093, Turkey;
| | - Rathan M. Subramaniam
- Dean’s Office, Otago Medical School, University of Otago, Dunedin 9054, New Zealand;
| | - Ayse Tuba Kendi
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
29
|
Kipnis ST, Hung M, Kumar S, Heckert JM, Lee H, Bennett B, Soulen MC, Pryma DA, Mankoff DA, Metz DC, Eads JR, Katona BW. Laboratory, Clinical, and Survival Outcomes Associated With Peptide Receptor Radionuclide Therapy in Patients With Gastroenteropancreatic Neuroendocrine Tumors. JAMA Netw Open 2021; 4:e212274. [PMID: 33755166 PMCID: PMC7988364 DOI: 10.1001/jamanetworkopen.2021.2274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE Peptide receptor radionuclide therapy (PRRT) is approved in the US for treatment of gastroenteropancreatic neuroendocrine tumors (NETs), but data on PRRT outcomes within US populations remain scarce. OBJECTIVE To analyze the first 2 years of PRRT implementation at a US-based NET referral center. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted using medical records of patients with metastatic NET receiving PRRT from 2018 through 2020 in a NET program at a tertiary referral center. Included patients were those at the center with metastatic NETs who received at least 1 dose of PRRT over the study period. Laboratory toxic effects were assessed using Common Terminology Criteria for Adverse Events version 5.0. Tumor response was determined using Response Evaluation Criteria in Solid Tumors 1.1. Survival analysis was conducted to identify factors associated with progression-free survival (PFS) and overall survival. Data were analyzed from August 2018 through August 2020. EXPOSURES Receiving 4 cycles of lutetium-177-dotatate infusion, separated by 8-week intervals targeted to 7.4 GBq (200 mCi) per dose. MAIN OUTCOMES AND MEASURES Data were compared from before and after PRRT to determine hematologic, liver, and kidney toxic effects and to assess tumor progression and patient survival. RESULTS Among 78 patients receiving at least 1 dose of PRRT, median (interquartile range) age at PRRT initiation was 59.8 (53.5-69.2) years and 39 (50.0%) were men. The most common primary NET sites included small bowel, occurring in 34 patients (43.6%), and pancreas, occurring in 22 patients (28.2%). World Health Organization grade 1 or 2 tumors occurred in 62 patients (79.5%). Among all patients, 56 patients underwent pretreatment with tumor resection (71.8%), 49 patients received nonsomatostatin analogue systemic therapy (62.8%), and 49 patients received liver-directed therapy (62.8%). At least 1 grade 2 or greater toxic effect was found in 47 patients (60.3%). Median PFS was 21.6 months for the study group, was not reached by 22 months for patients with small bowel primary tumors, and was 13.3 months for patients with pancreatic primary tumors. Having a small bowel primary tumor was associated with a lower rate of progression compared with having a pancreatic primary tumor (hazard ratio, 0.19; 95% CI, 0.07-0.55; P = .01). Median overall survival was not reached. CONCLUSIONS AND RELEVANCE This cohort study of patients with metastatic NETs found that PRRT was associated with laboratory-measured toxic effects during treatment for most patients and an overall median PFS of 21.6 months. Patients with small bowel NETs had longer PFS after PRRT compared with patients with pancreatic NETs.
Collapse
Affiliation(s)
- Sarit T. Kipnis
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Matthew Hung
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Shria Kumar
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jason M. Heckert
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia
| | - Hwan Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Bonita Bennett
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Michael C. Soulen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel A. Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David A. Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David C. Metz
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jennifer R. Eads
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Bryson W. Katona
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
30
|
Burkett BJ, Dundar A, Young JR, Packard AT, Johnson GB, Halfdanarson TR, Eiring RA, Gansen DN, Patton CM, Kendi AT. How We Do It: A Multidisciplinary Approach to 177Lu DOTATATE Peptide Receptor Radionuclide Therapy. Radiology 2020; 298:261-274. [PMID: 33231532 DOI: 10.1148/radiol.2020201745] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lutetium 177 (177Lu) DOTA-0-Tyr3-Octreotate (DOTATATE) peptide receptor radionuclide therapy (PRRT) is an effective treatment for advanced gastroenteropancreatic neuroendocrine tumors. This review presents a clinical practice workflow that has been successful since 177Lu DOTATATE PRRT was approved by the U.S. Food and Drug Administration. The workflow relies heavily on the input of a multidisciplinary team and involves a nuclear medicine consultation service, tumor board, and specific preparations in advance of therapy and day-of-therapy procedures. A systematic checklist designed to ensure appropriate selection of treatment candidates and identification of any concerns to address to safely administer PRRT is provided. All patients were evaluated with gallium 68 DOTATATE PET/CT, and in cases of high-grade tumors, they were also evaluated with fluorine 18 fluorodeoxyglucose PET/CT, with imaging findings reviewed as part of the systematic checklist before PRRT. Adverse effects are discussed and imaging follow-up regimens are reviewed, including alternative diagnostic contrast materials. Approaches to multiple challenging patient scenarios are illustrated through case examples. Finally, alternative theranostic radionuclides and treatment strategies are discussed.
Collapse
Affiliation(s)
- Brian J Burkett
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Ayca Dundar
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Jason R Young
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Annie T Packard
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Geoffrey B Johnson
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Thorvardur R Halfdanarson
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Rachel A Eiring
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Denise N Gansen
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Cynthia M Patton
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - A Tuba Kendi
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| |
Collapse
|
31
|
Abou Jokh Casas E, Pubul Núñez V, Anido-Herranz U, del Carmen Mallón Araujo M, del Carmen Pombo Pasín M, Garrido Pumar M, Cabezas Agrícola JM, Cameselle-Teijeiro JM, Hilal A, Ruibal Morell Á. Evaluation of 177Lu-Dotatate treatment in patients with metastatic neuroendocrine tumors and prognostic factors. World J Gastroenterol 2020; 26:1513-1524. [PMID: 32308351 PMCID: PMC7152518 DOI: 10.3748/wjg.v26.i13.1513] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/06/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND 177Lu peptide receptor radionuclide therapy (PRRT) is a recently approved therapy in Spain that has been demonstrated to be a well-tolerated therapy for positive somatostatin receptor advanced gastroenteropancreatic neuroendocrine tumors.
AIM To determine the impact of PRRT on quality of life, radiologic and metabolic response, overall survival, prognostic factors and toxicity.
METHODS Thirty-six patients treated with 177Lu-PRRT from 2016 to 2019 were included. The most frequent location of the primary tumor was the gastrointestinal tract (52.8%), pancreas (27.8%), and nongastropancreatic neuroendocrine tumor (11.1%). The liver was the most common site of metastasis (91.7%), followed by distant nodes (50.0%), bone (27.8%), peritoneum (25.0%) and lung (11.1%). Toxicity was evaluated after the administration of each dose. Treatment efficacy was evaluated by two parameters: stable disease and disease progression in response evaluation criteria in solid tumors 1.1 criterion and prognostic factors were tested.
RESULTS From 36 patients, 55.6% were men, with a median age of 61.1 ± 11.8 years. Regarding previous treatments, 55.6% of patients underwent surgery of the primary tumor, 100% of patients were treated with long-acting somatostatin analogues, 66.7% of patients were treated with everolimus, 27.8% of patients were treated with tyrosine kinase inhibitor, and 27.8% of patients were treated with interferon. One patient received radioembolization, three patients received chemoembolization, six patients received chemotherapy. Hematological toxicity was registered in 14 patients (G1-G2: 55.5% and G3: 3.1%). Other events presented were intestinal suboclusion in 4 cases, cholestasis in 2 cases and carcinoid crisis in 1 case. The median follow-up time was 3 years. Currently, 24 patients completed treatment. Nineteen are alive with stable disease, two have disease progression, eight have died, and nine are still receiving treatment. The median overall survival was 12.5 mo (95% confidence interval range: 9.8–15.2), being inversely proportional to toxicity in previous treatments (P < 0.02), tumor grade (P < 0.01) and the presence of bone lesions (P = 0.009) and directly proportional with matching lesion findings between Octreoscan and computed tomography pre-PRRT (P < 0.01), , primary tumor surgery (P = 0.03) and metastasis surgery (P = 0.045). In a multivariate Cox regression analysis, a high Ki67 index (P = 0.003), a mismatch in the lesion findings between Octreoscan and computed tomography pre-PRRT (P < 0.01) and a preceding toxicity in previous treatments (P < 0.05) were risk factors to overall survival.
CONCLUSION Overall survival was inversely proportional to previous toxicity, tumor grade and the presence of bone metastasis and directly proportional to matching lesion findings between Octreoscan and computed tomography pre-PRRT and primary tumor and metastasis surgery.
Collapse
Affiliation(s)
- Estephany Abou Jokh Casas
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Virginia Pubul Núñez
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Urbano Anido-Herranz
- Department of Oncology, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - María del Carmen Mallón Araujo
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Maria del Carmen Pombo Pasín
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - Miguel Garrido Pumar
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| | - José Manuel Cabezas Agrícola
- Department of Endocrinology, Santiago de Compostela´s University Hospital, Santiago de Compostela, 15706, A Coruña, Spain
| | | | - Ashraf Hilal
- Department of Statistics, University of Santiago de Compostela, Santiago de Compostela 15706, A Coruña, Spain
| | - Álvaro Ruibal Morell
- Department of Nuclear Medicine, Santiago de Compostela´s University Hospital, Santiago de Compostela 15706, A Coruña, Spain
| |
Collapse
|
32
|
Kendi AT, Mailman JA, Naraev BG, Mercer DJ, Underwood JK, Halfdanarson TR. Patient Travel Concerns After Treatment with 177Lu-DOTATATE. J Nucl Med 2020; 61:496-497. [PMID: 32169915 DOI: 10.2967/jnumed.120.243238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/15/2020] [Indexed: 11/16/2022] Open
Affiliation(s)
- Ayşe Tuba Kendi
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | |
Collapse
|
33
|
Patient Selection and Toxicities of PRRT for Metastatic Neuroendocrine Tumors and Research Opportunities. Curr Treat Options Oncol 2020; 21:25. [PMID: 32172368 DOI: 10.1007/s11864-020-0711-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OPINION STATEMENT Neuroendocrine tumors (NETs) are a heterogenous group of neoplasms characterized by varied biological hallmarks and behavior, ranging from indolent to aggressive. For many decades, somatostatin analogues and few targeted therapies were available for NETs and these therapies had minimal response rates. However, there have been a number of recent treatment advances. Peptide receptor radionuclide therapy (PRRT) is a novel approach to treatment of NETs and has changed the landscape of treatment for NETs. It is a form of targeted therapy in which a radiolabeled somatostatin analogue delivers radiation specifically to tumor cells expressing the somatostatin receptor.
Collapse
|
34
|
McGarrah PW, Westin GFM, Hobday TJ, Scales JA, Ingimarsson JP, Leibovich BC, Halfdanarson TR. Renal Neuroendocrine Neoplasms: A Single-center Experience. Clin Genitourin Cancer 2019; 18:e343-e349. [PMID: 31911122 DOI: 10.1016/j.clgc.2019.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Primary neuroendocrine neoplasms (NENs) of the kidney are exceedingly rare malignancies and the available literature is very limited. The natural history and response to treatments is not well characterized. We aimed to describe the presenting features, demographics, tumor characteristics, and treatment outcomes of patients with renal NENs. PATIENTS AND METHODS We performed a retrospective analysis of all Mayo Clinic patient records with a tissue diagnosis of a primary renal NEN. Baseline patient and surgical pathologic features and treatment modalities were collected. Time to recurrence after resection and overall survival (OS) were estimated using with survival analysis. Surveillance, Epidemiology, and End Results data were used to estimate the population-wide incidence and OS. RESULTS A total of 17 patients were included in the present study, with a median follow-up of 62.8 months. Distant metastasis was present in 29% at diagnosis, with 76% experiencing distant metastasis at any point; 24% had a horseshoe kidney. Of the 17 patients, 14 had undergone surgical resection with no evidence of disease postoperatively. Ten of these patients had documented recurrence. The median time to recurrence was 18 months (95% confidence interval, 9-46 months). Only 1 of the 10 patients showed a radiographic response to systemic therapy. Of 9 patients, 4 had stable disease with somatostatin analogs. The median OS was 143 months (95% confidence interval, 50-143 months). CONCLUSIONS Renal NENs are rare malignancies affecting mostly middle-age patients, with distant metastasis being common. Approximately one half of patients experience stable disease with somatostatin analogs. The OS usually exceeds 5 years.
Collapse
Affiliation(s)
| | - Gustavo F M Westin
- Division of Medical Oncology, Mayo Clinic, Rochester, MN; University Cancer and Blood Center, Athens, GA
| | | | - Joseph A Scales
- Department of Urology, Mayo Clinic, Rochester, MN; Urology Clinics of North Texas, Plano, TX
| | - Johann P Ingimarsson
- Department of Urology, Mayo Clinic, Rochester, MN; Maine Medical Partners Urology, South Portland, ME
| | | | | |
Collapse
|