1
|
Pavel M, Caplin ME, Ruszniewski P, Hertelendi M, Krenning EP, Strosberg JR. Relationship Between Best Tumor Shrinkage and Progression-Free Survival and Overall Survival in Patients With Progressive Midgut Neuroendocrine Tumors Treated With [ 177Lu]Lu-DOTA-TATE: Ad Hoc Analysis of the Phase III NETTER-1 Trial. Cancer Med 2025; 14:e70744. [PMID: 40272146 PMCID: PMC12020026 DOI: 10.1002/cam4.70744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND In many solid tumors, early tumor shrinkage predicts the durability of treatment response. It is unclear whether this is the case for neuroendocrine tumors treated with peptide receptor radionuclide therapy (PRRT). METHODS Data from the phase III NETTER-1 study of [177Lu]Lu-DOTA-TATE (177Lu-DOTATATE) for the treatment of advanced, well-differentiated, midgut NETs were used to investigate whether objective tumor shrinkage (local review) with 177Lu-DOTATATE is associated with progression-free survival (PFS) and overall survival (OS) duration. RESULTS Overall, 117 patients were treated with 177Lu-DOTATATE (four cycles of 7.4 GBq every 8 weeks). In a landmark analysis, best tumor shrinkage from baseline until data cut-off (prior to first progression) was not associated with PFS (n = 102; hazard ratio: 1.002 [95% confidence interval (CI): 0.99-1.02]; nominal p = 0.7808). In further ad hoc analyses, patients on the 177Lu-DOTATATE arm were dichotomized into ≥ 30% tumor shrinkage from baseline (18/117 [15.4%]) and < 30% shrinkage (99/117 [84.6%]). Median (95% CI) PFS was 17.6 (16.5-30.3) months in the ≥ 30% shrinkage group and 25.0 (19.4-31.0) months in the < 30% group. OS was not significantly different for the two tumor shrinkage groups (not estimable [31.0 months-not estimable] and 44.3 [34.9-53.8] months, respectively). CONCLUSIONS These results suggest the benefit of PRRT and the potential PFS and OS benefit of 177Lu-DOTATATE should not be based on tumor shrinkage (objective response versus stable disease) and that lack of tumor shrinkage should not impact application of the approved four cycles of 177Lu-DOTATATE.
Collapse
Affiliation(s)
- Marianne Pavel
- Department of Medicine 1, Uniklinikum Erlangen and Comprehensive Cancer Center CCC‐EMNFriedrich Alexander University Erlangen‐NürnbergErlangenGermany
| | | | - Philippe Ruszniewski
- Université Paris Cité and Department of Pancreatology and Digestive Oncology, Beaujon HospitalClichyFrance
| | | | - Eric P. Krenning
- Cyclotron Rotterdam B.V., Erasmus Medical CenterRotterdamthe Netherlands
| | | | | |
Collapse
|
2
|
Zhao J, Bera K, Mohamed A, Li Q, Ramaiya N, Tirumani SH. Comparison of RECIST 1.1, mRECIST and PERCIST for assessment of peptide receptor radionuclide therapy treatment response in metastatic neuroendocrine tumors. Curr Probl Diagn Radiol 2025; 54:228-232. [PMID: 39389807 DOI: 10.1067/j.cpradiol.2024.10.003] [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: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE To compare RECIST 1.1, modified RECIST (mRECIST) and PERCIST for assessment of Peptide Receptor Radionuclide Therapy (PRRT) treatment response in metastatic neuroendocrine tumors. MATERIALS In this IRB-approved, HIPAA compliant retrospective study, patients treated with PRRT between July 2019 and Dec 2022 were identified. Inclusion criteria were presence of at least one pre-and one post-treatment imaging (CT, MRI, Ga 68 or Cu64 DOTATATE PET/CT) within one year of the start and end of PRRT respectively. The imaging was reviewed independently by two radiologists using RECIST 1.1, modified RECIST (mRECIST) and PERCIST criteria. Response of first post treatment scan and presence of disease progression during follow-up were recorded along with the date of best response and disease progression. Statistical analysis was performed to determine inter-reader agreement and agreement between the various response criteria using kappa statistics. RESULTS Best response by RECIST 1.1 was recorded in 26 patients (PR-7, SD- 13, PD- 6), by mRECIST in 22 patients (PR-7, SD- 10, PD- 5), by PERCIST in 14 patients (PR-4, SD- 3, PD- 7). Inter-reader agreement was highest for PERCIST (weighted kappa 0.921, standard error 0.078 95% CI 0.769 to 1.000) followed by RECIST 1.1 (weighted kappa 0.897, standard error 0.071 95% CI 0.758 to 1.000) and mRECIST (weighted kappa 0.883, standard error 0.079 95% CI 0.727 to 1.000).
Collapse
Affiliation(s)
- Jack Zhao
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Kaustav Bera
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.
| | - Amr Mohamed
- Medical Oncology, Department of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Qiubai Li
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nikhil Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
3
|
Liu X, Han X, Zhang G, Zhu X, Zhang W, Wang X, Wu C. Computed tomography-based delta-radiomics analysis for preoperative prediction of ISUP pathological nuclear grading in clear cell renal cell carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04857-4. [PMID: 40024922 DOI: 10.1007/s00261-025-04857-4] [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: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Nuclear grading of clear cell renal cell carcinoma (ccRCC) plays a crucial role in diagnosing and managing the disease. OBJECTIVE To develop and validate a CT-based Delta-Radiomics model for preoperative assessment of nuclear grading in renal clear cell carcinoma. MATERIALS AND METHODS This retrospective analysis included surgical cases of 146 ccRCC patients from two medical centers from December 2018 to December 2023, with 117 patients from Hospital and 29 from the *Hospital Affiliated to University of **. Radiomic features were extracted from whole-abdomen CT images, and the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm was used for feature selection. The Multi-Layer Perceptron (MLP) approach was employed to construct five predictive models (RAD_NE, RAD_AP, RAD_VP, RAD_Delta1, RAD_Delta2). The models were evaluated using area under the curve (AUC), accuracy, sensitivity, and specificity, while clinical utility was assessed through Decision Curve Analysis (DCA). RESULTS A total of 1,834 radiomic features were extracted from the three phases of the CT images for each model. The models demonstrated strong classification performance, with AUC values ranging from 0.837 to 0.911 in the training set and 0.608 to 0.869 in the test set. The Rad_Delta1 and Rad_Delta2 models demonstrated advantages in predicting ccRCC pathological grading.The AUC value of the Rad_Delta1 is 0.911in the training set and 0.771 in the external verifcation set.The AUC value of the Rad_Delta2 is 0.881 in the training set and0.608 in the external verifcation set. DCA curves confirmed the clinical applicability of these models. CONCLUSION CT-based delta-radiomics shows potential in predicting the pathological grading of clear cell renal cell carcinoma (ccRCC).
Collapse
Affiliation(s)
- Xiaohui Liu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiaowei Han
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| | - Guozheng Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| | - Xisong Zhu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Wen Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xu Wang
- The Afliated Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Chenghao Wu
- Quzhou Architectural Design Institute Co., Ltd, Quzhou, China
| |
Collapse
|
4
|
Kong G, Noe G, Chiang C, Herrmann K, Hope TA, Michael M. Assessment of response to PRRT including anatomical and molecular imaging as well as novel biomarkers. J Neuroendocrinol 2025; 37:e13461. [PMID: 39520276 PMCID: PMC11919480 DOI: 10.1111/jne.13461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/05/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) is an effective treatment for both oncological and hormone control and is a widely accepted standard of care treatment for patients with neuroendocrine neoplasms (NEN). Its use is anticipated to increase significantly, and this demands accurate tools and paradigms to assess treatment response post PRRT. This article outlines the current role and future developments of anatomical, molecular imaging and biomarkers for response assessment to PRRT, highlighting the challenges and provides perspectives for the need to focus on a multimodality, multidisciplinary and individualised approach for patients with this complex heterogeneous disease.
Collapse
Affiliation(s)
- Grace Kong
- Department of Molecular Imaging and Therapeutic Nuclear MedicinePeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Geertje Noe
- Department of Cancer ImagingPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Cherie Chiang
- Department of Internal MedicinePeter MacCallum Cancer CentreParkvilleVictoriaAustralia
- Department of Diabetes and Endocrinology, Melbourne HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Ken Herrmann
- Department of Nuclear MedicineUniversity of Duisburg‐Essen and German Cancer Consortium (DKTK)‐University Hospital EssenEssenGermany
| | - Thomas A. Hope
- Department of RadiologySan Francisco VA Medical CenterSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Michael Michael
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| |
Collapse
|
5
|
Prasad V, Koumarianou A, Denecke T, Sundin A, Deroose CM, Pavel M, Christ E, Lamarca A, Caplin M, Castaño JP, Dromain C, Falconi M, Grozinsky-Glasberg S, Hofland J, Knigge UP, Kos-Kudla B, Krishna BA, Reed NS, Scarpa A, Srirajaskanthan R, Toumpanakis C, Kjaer A, Hicks RJ, Ambrosini V. Challenges in developing response evaluation criteria for peptide receptor radionuclide therapy: A consensus report from the European Neuroendocrine Tumor Society Advisory Board Meeting 2022 and the ENETS Theranostics Task Force. J Neuroendocrinol 2025; 37:e13479. [PMID: 39653582 DOI: 10.1111/jne.13479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025]
Abstract
Assessing the response to systemic therapy in neuroendocrine tumors (NET) is challenging since morphological imaging response is often delayed and not necessarily reflective of clinical benefit. Peptide receptor radionuclide therapy (PRRT) has a complex mechanism of action, further complicating response assessment. In response to these challenges, the European Neuroendocrine Tumor Society (ENETS) Theranostics Task Force conducted a statement-based survey among experts to identify the current landscape and unmet needs in PRRT response assessment. The survey, presented at the 2022 ENETS Advisory Board (AB) meeting in Vienna, was completed by 70% of AB members, most of whom (81%) were from ENETS Centers of Excellence (CoE). It comprised a set of 13 questions with two substatements in three questions. Six (46%) of the statements achieved more than 75% agreement, while five (39%) additional statements reached over 60% consensus. Key points from the survey include: AB members agreed that lesions deemed equivocal on computed tomography (CT) or magnetic resonance imaging (MRI) should be characterized by somatostatin receptor (SST) positron emission tomography (PET)/CT before being designated as target lesions. It was agreed that interim response assessments should occur after the second or third PRRT cycle. Over half (54%) preferred using both conventional cross-sectional imaging (CT and/or MRI) and hybrid imaging (SST PET/CT) for this purpose. Almost all AB members supported further response assessment 3 months after the final PRRT cycle. A majority (62%) preferred using a combination of conventional cross-sectional imaging and SST PET/CT. For cases showing equivocal progression (ambiguous lesions or nontarget lesions) on CT and/or MRI, further confirmation using SST PET/CT was recommended. A significant majority (74%) preferred assessing pseudo-progression and delayed response by combining SST PET with diagnostic CT and/ or MRI. Though just below the 75% consensus threshold, there was substantial agreement on selecting target lesions based on SST PET/CT uptake intensity and homogeneity. Sixty-nine percent noted the importance of documenting and closely following heterogeneity in lesions in liver, lymph nodes, primary tumors, or other organs. As to the statement on parameters for new response criteria, AB members recommended exploring maximum standard unit value, tumor-to-background ratio, Hounsfield Unit (Choi Criteria), total tumor burden, and novel serum or molecular markers for future response evaluation criteria. Sixty-five percent supported the use of a single SST PET/CT for response assessment of NET lesions treated with PRRT. These findings highlight the importance of integrating advanced imaging techniques and recognizing the need for more nuanced criteria in assessing the efficacy of PRRT in NET patients. This approach aims to enhance the accuracy of treatment monitoring and improve patient outcomes.
Collapse
Affiliation(s)
- Vikas Prasad
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, USA
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, University Medical Centre Leipzig, Leipzig, Germany
| | - Anders Sundin
- Radiology and Molecular Imaging, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals Leuven and Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Marianne Pavel
- Department of Medicine 1, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Emanuel Christ
- Center of Endocrine and Neuroendocrine Tumors, ENETS Center of Excellence (CoE), Division of Endocrinology, Diabetology and Metabolism, University Hospital of Basel, Basel, Switzerland
| | - Angela Lamarca
- Department of Oncology-OncoHealth Institute-Instituto de Investigaciones Sanitarias FJD, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Martyn Caplin
- Neuroendocrine Tumour Unit, ENETS Center of Excellence, Royal Free Hospital, London, UK
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Córdoba, Spain
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Massimo Falconi
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Simona Grozinsky-Glasberg
- Neuroendocrine Tumor Unit, ENETS Center of Excellence, Division of Medicine, Hadassah Medical Organization and Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Johannes Hofland
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ulrich Peter Knigge
- Department of Surgery and Transplantation, Department of Nephrology and Endocrinology, Center of Cancer and Transplantation, ENETS Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Beata Kos-Kudla
- Department of Endocrinology and Neuroendocrine Tumours, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
| | - Balkundi A Krishna
- Department of Nuclear Medicine & PET imaging, Lilavati Hospital & Research Centre, Mumbai, India
| | | | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Christos Toumpanakis
- Neuroendocrine Tumour Unit, ENETS Center of Excellence, Royal Free Hospital, London, UK
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rodney J Hicks
- St Vincent's Hospital, Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Translational Medicine, the Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
6
|
Karim H, Winkelmann M, Grawe F, Völter F, Auernhammer C, Rübenthaler J, Ricke J, Ingenerf M, Schmid-Tannwald C. Quantitative SSTR-PET/CT: a potential tool for predicting everolimus response in neuroendoctine tumour patients. Radiol Oncol 2024; 58:348-356. [PMID: 38861687 PMCID: PMC11406901 DOI: 10.2478/raon-2024-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND This study aimed to assess 68Ga-DOTA-TATE (-TOC) PET/CT quantitative parameters in monitoring and predicting everolimus response in neuroendocrine tumor (NET) patients with hepatic metastases (NELM). PATIENTS AND METHODS This retrospective analysis included 29 patients with 62 target lesions undergoing everolimus treatment and pre-therapy, and follow-up 68Ga-DOTA-TATE (-TOC) PET/CT scans. Response evaluation utilized progression-free survival (PFS) categorized as responders (R; PFS > 6 months) and non-responders (NR; PFS ≤ 6 months). Lesion size and density, along with maximum and median standardize uptake value (SUV) in target lesions, liver, and spleen were assessed. Tumor-to-spleen (T/S) and tumor-to-liver (T/L) ratios were calculated, including the tumor-to-spleen (T/S) ratio and tumor-to-liver (T/L) ratio (using SUVmax/SUVmax, SUVmax/SUVmean, and SUVmean/SUVmean). RESULTS PET/CT scans were acquired 19 days (interquartile range [IQR] 69 days) pre-treatment and 127 days (IQR 74 days) post-starting everolimus. The overall median PFS was 264 days (95% CI: 134-394 days). R exhibited significant decreases in Tmax/Lmax and Tmean/Lmax ratios compared to NR (p = 0.01). In univariate Cox regression, Tmean/Lmax ratio was the sole prognostic parameter associated with PFS (HR 0.5, 95% CI 0.28-0.92, p = 0.03). Percentage changes in T/L and T/S ratios were significant predictors of PFS, with the highest area under curve (AUC) for the percentage change of Tmean/Lmax (AUC = 0.73). An optimal threshold of < 2.5% identified patients with longer PFS (p = 0.003). No other imaging or clinical parameters were predictive of PFS. CONCLUSIONS This study highlights the potential of quantitative SSTR-PET/CT in predicting and monitoring everolimus response in NET patients. Liver metastasis-to-liver parenchyma ratios outperformed size-based criteria, and Tmean/Lmax ratio may serve as a prognostic marker for PFS, warranting larger cohort investigation.
Collapse
Affiliation(s)
- Homeira Karim
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Freba Grawe
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Friederike Völter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Auernhammer
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
- Department of Internal Medicine 4, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Rübenthaler
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
| | - Maria Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Christine Schmid-Tannwald
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
| |
Collapse
|
7
|
Gao J, Zhou J, Liu C, Pan Y, Lin X, Zhang Y. Outcome prediction of SSTR-RADS-3A and SSTR-RADS-3B lesions in patients with neuroendocrine tumors based on 68Ga-DOTATATE PET/MR. J Cancer Res Clin Oncol 2024; 150:272. [PMID: 38795250 PMCID: PMC11127844 DOI: 10.1007/s00432-024-05776-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 05/27/2024]
Abstract
PURPOSE Somatostatin receptor (SSTR)-targeted PET imaging has emerged as a common approach to evaluating those patients with well-differentiated neuroendocrine tumors (NETs). The SSTR reporting and data system (SSTR-RADS) version 1.0 provides a means of categorizing lesions from 1 to 5 according to the likelihood of NET involvement, with SSTR-RADS-3A (soft-tissue) and SSTR-RADS-3B (bone) lesions being those suggestive of but without definitive NET involvement. The goal of the present study was to assess the ability of 68Ga-DOTATATE PET/MR imaging data to predict outcomes for indeterminate SSTR-RADS-3A and 3B lesions. METHODS NET patients with indeterminate SSTR-RADS-3A or SSTR-RADS-3B lesions who underwent 68Ga-DOTATATE PET/MR imaging from April 2020 through August 2023 were retrospectively evaluated. All patients underwent follow-up through December 2023 (median, 17 months; (3-31 months)), with imaging follow-up or biopsy findings ultimately being used to classify lesions as malignant or benign. Lesion maximum standardized uptake value (SUVmax) along with minimum and mean apparent diffusion coefficient (ADCmin and ADCmean) values were measured and assessed for correlations with outcomes on follow-up. RESULTS In total, 33 indeterminate SSTR-RADS-3 lesions from 22 patients (19 SSTR-RADS-3A and 14 SSTR-RADS-3B) were identified based upon baseline 68Ga-DOTATATE PET/MR findings. Over the course of follow-up, 16 of these lesions (48.5%) were found to exhibit true NET positivity, including 9 SSTR-RADS-3A and 7 SSTR-RADS-3B lesions. For SSTR-RADS-3A lymph nodes, a diameter larger than 0.7 cm and an ADCmin of 779 × 10-6mm2/s or lower were identified as being more likely to be associated with metastatic lesions. Significant differences in ADCmin and ADCmean were identified when comparing metastatic and non-metastatic SSTR-RADS-3B bone lesions (P < 0.05), with these parameters offering a high predictive ability (AUC = 0.94, AUC = 0.86). CONCLUSION Both diameter and ADCmin can aid in the accurate identification of the nature of lesions associated with SSTR-RADS-3A lymph nodes, whereas ADCmin and ADCmean values can inform the accurate interpretation of SSTR-RADS-3B bone lesions.
Collapse
Affiliation(s)
- Jing Gao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Jinxin Zhou
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Chang Liu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yu Pan
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Xiaozhu Lin
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China.
| | - Yifan Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
8
|
Ingenerf M, Karim H, Auernhammer C, Zacherl M, Wenter V, Winkelmann M, Ricke J, Berger F, Schmid-Tannwald C. Quantitative SSTR-PET/CT for predicting response and survival outcomes in patients with pancreatic neuroendocrine tumors receiving CAPTEM. Radiol Oncol 2023; 57:436-445. [PMID: 38038419 PMCID: PMC10690751 DOI: 10.2478/raon-2023-0055] [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: 06/05/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the predictive and monitoring role of somatostatin receptor (SSTR) positron emission tomography-computed tomography (PET/CT) and clinical parameters in patients with neuroendocrine liver metastases (NELM) from pancreatic neuroendocrine tumors (pNET) receiving capecitabine and temozolomide (CAPTEM). PATIENTS AND METHODS This retrospective study included twenty-two patients with pNET and NELM receiving CAPTEM who underwent pre- and post-therapeutic 68Ga-DOTATATE/-TOC PET/CT. Imaging (including standardized uptake value [SUV] of target lesions [NELM and pNET], normal spleen and liver) and clinical (Chromogranin A [CgA], Ki-67) parameters were assessed. Treatment outcome was evaluated as response according to RECIST 1.1, progression free survival (PFS) and overall survival (OS). RESULTS The median PFS (mPFS) was 7 months. Responders had a significantly longer mPFS compared to non-responders (10 vs. 4 months p = 0.022). Median OS (mOS) was 33 months (mOS: responders = 80 months, non-responders = 24 months p = 0.182). Baseline imaging showed higher SUV in responders, including absolute SUV, tumor-to-spleen (T/S), and tumor-to-liver (T/L) ratios (p < 0.02). All SUV parameters changed only in the responders during follow-up. Univariable Cox regression analysis identified baseline Tmax/Smean ratio and percentage change in size of pNETs as significant factors associated with PFS. A baseline Tmax/Smean ratio < 1.5 was associated with a shorter mPFS (10 vs. 4 months, (p < 0.05)). Prognostic factors for OS included age, percentage change in CgA and in T/S ratios in univariable Cox regression. CONCLUSIONS SSTR-PET/CT can be useful for predicting response and survival outcomes in pNET patients receiving CAPTEM: Higher baseline SUV values, particularly Tmax/Smean ratios of liver metastases were associated with better response and prolonged PFS.
Collapse
Affiliation(s)
- Maria Ingenerf
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Homeira Karim
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Auernhammer
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
- Department of Internal Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Zacherl
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Vera Wenter
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Michael Winkelmann
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
| | - Frank Berger
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Christine Schmid-Tannwald
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- ENETS Centre of Excellence, Interdisciplinary Center of Neuroendocrine Tumours of the GastroEnteroPancreatic System at the University Hospital of Munich (GEPNET-KUM), University Hospital of Munich, Munich, Germany
| |
Collapse
|
9
|
Gabiache G, Zadro C, Rozenblum L, Vezzosi D, Mouly C, Thoulouzan M, Guimbaud R, Otal P, Dierickx L, Rousseau H, Trepanier C, Dercle L, Mokrane FZ. Image-Guided Precision Medicine in the Diagnosis and Treatment of Pheochromocytomas and Paragangliomas. Cancers (Basel) 2023; 15:4666. [PMID: 37760633 PMCID: PMC10526298 DOI: 10.3390/cancers15184666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
In this comprehensive review, we aimed to discuss the current state-of-the-art medical imaging for pheochromocytomas and paragangliomas (PPGLs) diagnosis and treatment. Despite major medical improvements, PPGLs, as with other neuroendocrine tumors (NETs), leave clinicians facing several challenges; their inherent particularities and their diagnosis and treatment pose several challenges for clinicians due to their inherent complexity, and they require management by multidisciplinary teams. The conventional concepts of medical imaging are currently undergoing a paradigm shift, thanks to developments in radiomic and metabolic imaging. However, despite active research, clinical relevance of these new parameters remains unclear, and further multicentric studies are needed in order to validate and increase widespread use and integration in clinical routine. Use of AI in PPGLs may detect changes in tumor phenotype that precede classical medical imaging biomarkers, such as shape, texture, and size. Since PPGLs are rare, slow-growing, and heterogeneous, multicentric collaboration will be necessary to have enough data in order to develop new PPGL biomarkers. In this nonsystematic review, our aim is to present an exhaustive pedagogical tool based on real-world cases, dedicated to physicians dealing with PPGLs, augmented by perspectives of artificial intelligence and big data.
Collapse
Affiliation(s)
- Gildas Gabiache
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Charline Zadro
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Laura Rozenblum
- Department of Nuclear Medicine, Sorbonne Université, AP-HP, Hôpital La Pitié-Salpêtrière, 75013 Paris, France
| | - Delphine Vezzosi
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | - Céline Mouly
- Department of Endocrinology, Rangueil University Hospital, 31400 Toulouse, France
| | | | - Rosine Guimbaud
- Department of Oncology, Rangueil University Hospital, 31400 Toulouse, France
| | - Philippe Otal
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Lawrence Dierickx
- Department of Nuclear Medicine, IUCT-Oncopole, 31059 Toulouse, France;
| | - Hervé Rousseau
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| | - Christopher Trepanier
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Laurent Dercle
- New York-Presbyterian Hospital/Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fatima-Zohra Mokrane
- Department of Radiology, Rangueil University Hospital, 31400 Toulouse, France (F.-Z.M.)
| |
Collapse
|
10
|
Mitjavila M, Jimenez-Fonseca P, Belló P, Pubul V, Percovich JC, Garcia-Burillo A, Hernando J, Arbizu J, Rodeño E, Estorch M, Llana B, Castellón M, García-Cañamaque L, Gajate P, Riesco MC, Miguel MB, Balaguer-Muñoz D, Custodio A, Cano JM, Repetto A, Garcia-Alonso P, Muros MA, Vercher-Conejero JL, Carmona-Bayonas A. Efficacy of [ 177Lu]Lu-DOTATATE in metastatic neuroendocrine neoplasms of different locations: data from the SEPTRALU study. Eur J Nucl Med Mol Imaging 2023; 50:2486-2500. [PMID: 36877234 PMCID: PMC10250456 DOI: 10.1007/s00259-023-06166-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/18/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Peptide receptor radionuclide therapy (PRRT) is one of the most promising therapeutic strategies in neuroendocrine neoplasms (NENs). Nevertheless, its role in certain tumor sites remains unclear. This study sought to elucidate the efficacy and safety of [177Lu]Lu-DOTATATE in NENs with different locations and evaluate the effect of the tumor origin, bearing in mind other prognostic variables. Advanced NENs overexpressing somatostatin receptors (SSTRs) on functional imaging, of any grade or location, treated at 24 centers were enrolled. The protocol consisted of four cycles of 177Lu-DOTATATE 7.4 GBq iv every 8 weeks (NCT04949282). RESULTS The sample comprised 522 subjects with pancreatic (35%), midgut (28%), bronchopulmonary (11%), pheochromocytoma/ paraganglioma (PPGL) (6%), other gastroenteropancreatic (GEP) (11%), and other non-gastroenteropancreatic (NGEP) (9%) NENs. The best RECIST 1.1 responses were complete response, 0.7%; partial response, 33.2%; stable disease, 52.1%; and tumor progression, 14%, with activity conditioned by the tumor subtype, but with benefit in all strata. Median progression-free survival (PFS) was 31.3 months (95% CI, 25.7-not reached [NR]) in midgut, 30.6 months (14.4-NR) in PPGL, 24.3 months (18.0-NR) in other GEP, 20.5 months (11.8-NR) in other NGEP, 19.8 months (16.8-28.1) in pancreatic, and 17.6 months (14.4-33.1) in bronchopulmonary NENs. [177Lu]Lu-DOTATATE exhibited scant severe toxicity. CONCLUSION This study confirms the efficacy and safety of [177Lu]Lu-DOTATATE in a wide range of SSTR-expressing NENs, regardless of location, with clinical benefit and superimposable survival outcomes between pNENs and other GEP and NGEP tumor subtypes different from midgut NENs.
Collapse
Affiliation(s)
- Mercedes Mitjavila
- Department of Nuclear Medicine, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Paula Jimenez-Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Pilar Belló
- Department of Nuclear Medicine, Hospital Universitario La Fe, Valencia, Spain
| | - Virginia Pubul
- Department of Nuclear Medicine, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Juan Carlos Percovich
- Department of Endocrinology and Nutrition, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Amparo Garcia-Burillo
- Department of Nuclear Medicine, Hospital Universitario Vall d’Hebron, Barcelona, Spain
| | - Jorge Hernando
- Department of Medical Oncology, Hospital Universitario Vall d’Hebron, Vall Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Javier Arbizu
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Emilia Rodeño
- Department of Nuclear Medicine, Hospital Universitario de Cruces, Bilbao, Spain
| | - Montserrat Estorch
- Department of Nuclear Medicine, Hospital de la Santa Creu i San Pau, Barcelona, Spain
| | - Belén Llana
- Department of Nuclear Medicine, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Maribel Castellón
- Department of Nuclear Medicine, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Pablo Gajate
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Maria Carmen Riesco
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria Begoña Miguel
- Department of Nuclear Medicine, Hospital Universitario de Burgos, Burgos, Spain
| | - David Balaguer-Muñoz
- Department of Nuclear Medicine, Hospital Universitario Doctor Peset, Valencia, Spain
| | - Ana Custodio
- Department of Medical Oncology, Hospital Universitario La Paz, CIBERONC CB16/12/00398, Madrid, Spain
| | - Juana María Cano
- Department of Medical Oncology, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Alexandra Repetto
- Department of Nuclear Medicine, Hospital Universitario Son Espases, Mallorca, Spain
| | - Pilar Garcia-Alonso
- Department of Nuclear Medicine, Hospital Universitario de Getafe, Madrid, Spain
| | - Maria Angustias Muros
- Department of Nuclear Medicine, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Jose Luis Vercher-Conejero
- Department of Nuclear Medicine-PET Unit, Hospital Universitario de Bellvitge - IDIBELL, Barcelona, Spain
| | - Alberto Carmona-Bayonas
- Department of Medical Oncology, Hospital Universitario Morales Meseguer, University de Murcia, IMIB, Murcia, Spain
| |
Collapse
|
11
|
Wnt/β-catenin modulating drugs regulate somatostatin receptor expression and internalization of radiolabelled octreotide in neuroendocrine tumor cells. Nucl Med Commun 2023; 44:259-269. [PMID: 36804512 DOI: 10.1097/mnm.0000000000001666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
BACKGROUND Differentiated neuroendocrine tumors (NETs) express somatostatin receptors (SSTRs), targets for therapy with either unlabeled or radioactively labeled somatostatin analogs (SSA). Associated with worse prognosis, dedifferentiated NET loose SSTR expression, which may be linked to deregulation of Wnt/β-catenin signaling on an intracellular level. The aim of the present study was to investigate the effect of Wnt/β-catenin signaling pathway alterations on SSTR expression and its function in NET. METHODS The NET cell lines BON-1 and QGP-1 were incubated with the Wnt-inhibitors 5-aza-2'-deoxycytidine (5-aza-CdR), Quercetin, or Niclosamide, or the Wnt activator lithium chloride (LiCl). Expression of SSTR1, SSTR2, and SSTR5 was determined by quantitative RT-PCR (qRT-PCR), immunocytomicroscopy and western blot. Changes in the Wnt pathway were analyzed by qRT-PCR of selected target genes and the TaqMan Array Human WNT Pathway. Receptor-associated function was determined by measuring the cellular uptake of [125I-Tyr3] octreotide. RESULTS The mRNAs of SSTRs 1-5 were expressed in both cell lines. Wnt inhibitors caused downregulation of Wnt target genes, while 5-aza-CdR had the highest inhibitory effect. LiCl lead to an upregulation of Wnt genes, which was more marked in QGP-1 cells. SSTR expression increased in both cell lines upon Wnt inhibition. All three Wnt inhibitors lead to a marked increase in the specific uptake of [125I-Tyr3]octreotide, with 5-aza-CdR showing the greatest effect (increase by more than 50% in BON-1 cells), while a decreased uptake of [125I-Tyr3]octreotide was seen upon activation of Wnt signaling by LiCl. CONCLUSIONS We demonstrate here that Wnt signaling orchestrates SSTR expression and function in a preclinical NET model. Wnt inhibition increases [125I-Tyr3]octreotide uptake offering an opportunity to enhance the efficacy of SSTR-targeted theranostic approaches.
Collapse
|
12
|
Parghane RV, Mahajan A, Chakrabarty N, Basu S. Imaging Recommendations for Theranostic PET-CT in Oncology. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1760310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
AbstractWe in this article have presented a review of the guideline recommendations on theranostic positron emission tomography-computed tomography (PET-CT) imaging which will be helpful to assist practitioners in providing appropriate patient care. Multiple guidelines by different societies and medical associations provide standards for diagnosis, imaging, and treatment of cancer patients. They have generated a number of recommendations related to 68Ga-DOTATATE and 68Ga-PSMA-11 PET-CT, which are the classical examples of theranostic PET-CT imaging in current practice.
Collapse
Affiliation(s)
- Rahul V. Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Abhishek Mahajan
- The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Place, Liverpool, United Kingdom
| | - Nivedita Chakrabarty
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
13
|
Veenstra EB, Noordzij W, Erba PA. Neuroendocrine Neoplasm Imaging and Image-Guided Therapies. MULTIMODALITY IMAGING AND INTERVENTION IN ONCOLOGY 2023:419-439. [DOI: 10.1007/978-3-031-28524-0_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Rajamohan N, Khasawneh H, Singh A, Suman G, Johnson GB, Majumder S, Halfdanarson TR, Goenka AH. PET/CT and PET/MRI in neuroendocrine neoplasms. Abdom Radiol (NY) 2022; 47:4058-4072. [PMID: 35426497 DOI: 10.1007/s00261-022-03516-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 01/18/2023]
Abstract
Advanced molecular imaging has come to play an integral role in the management of gastro-entero-pancreatic neuroendocrine neoplasms (GEP-NENs). Somatostatin receptor (SSTR) PET has now emerged as the reference standard for the evaluation of NENs and is particularly critical in the context of peptide receptor radionuclide therapy (PRRT) eligibility. SSTR PET/MRI with liver-specific contrast agent has a strong potential for one-stop-shop multiparametric evaluation of GEP-NENs. 18F-FDG is a complementary radiotracer to SSTR, especially in the context of high-grade neuroendocrine neoplasms. Knowledge gaps in quantitative evaluation of molecular imaging studies and their role in assessment of response to PRRT and combination therapies are active research areas. Novel radiotracers have the potential to overcome existing limitations in the molecular imaging of GEP-NENs. The purpose of this article is to provide an overview of the current trends, pitfalls, and recent advancements of molecular imaging for GEP-NENs.
Collapse
Affiliation(s)
- Naveen Rajamohan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Aparna Singh
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Garima Suman
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, 200 First Street SW, Charlton 1, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Treatment Assessment of pNET and NELM after Everolimus by Quantitative MRI Parameters. Biomedicines 2022; 10:biomedicines10102618. [PMID: 36289880 PMCID: PMC9599819 DOI: 10.3390/biomedicines10102618] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/01/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Assessment of treatment response to targeted therapies such as everolimus is difficult, especially in slow-growing tumors such as NETs. In this retrospective study, 17 patients with pancreatic neuroendocrine tumors (pNETs) and hepatic metastases (NELMs) (42 target lesions) who received everolimus were analyzed. Intralesional signal intensities (SI) of non-contrast T1w, T2w and DCE imaging, and apparent diffusion coefficients (ADCmean and ADCmin) of DWI, were measured on baseline and first follow-up MRI after everolimus initiation. Response assessment was categorized according to progression-free survival (PFS), with responders (R) showing a PFS of ≥11 months. ADCmin of NELMs decreased in Rs whereas it increased in non-responders (NR). Percentual changes of ADCmin and ADCmean differed significantly between response groups (p < 0.03). By contrast, ADC of the pNETs tended to increase in Rs, while there was no change in NRs. Tumor-to-liver (T/L) ratio of T1 SI of NELMs increased in Rs and decreased in NRs, and percentual changes differed significantly between response groups (p < 0.02). T1 SI of the pNETs tended to decrease in Rs and increase in Ns. The quotient of pretherapeutic and posttherapeutic ADCmin values (DADCmin) and length of everolimus treatment showed significant association with PFS in univariable Cox analysis. In conclusion, quantitative MRI, especially DWI, seems to allow treatment assessment of pNETs with NELMs under everolimus. Interestingly, the responding NELMs showed decreasing ADC values, and there might be an opposite effect on ADC and T1 SI between NELMs and pNETs.
Collapse
|
16
|
Bartolomei M, Berruti A, Falconi M, Fazio N, Ferone D, Lastoria S, Pappagallo G, Seregni E, Versari A. Clinical Management of Neuroendocrine Neoplasms in Clinical Practice: A Formal Consensus Exercise. Cancers (Basel) 2022; 14:2501. [PMID: 35626105 PMCID: PMC9140035 DOI: 10.3390/cancers14102501] [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: 05/03/2022] [Accepted: 05/16/2022] [Indexed: 02/01/2023] Open
Abstract
Many treatment approaches are now available for neuroendocrine neoplasms (NENs). While several societies have issued guidelines for diagnosis and treatment of NENs, there are still areas of controversy for which there is limited guidance. Expert opinion can thus be of support where firm recommendations are lacking. A group of experts met to formulate 14 statements relative to diagnosis and treatment of NENs and presented herein. The nominal group and estimate-talk-estimate techniques were used. The statements covered a broad range of topics from tools for diagnosis to follow-up, evaluation of response, treatment efficacy, therapeutic sequence, and watchful waiting. Initial prognostic characterization should be based on clinical information as well as histopathological analysis and morphological and functional imaging. It is also crucial to optimize RLT for patients with a NEN starting from accurate characterization of the patient and disease. Follow-up should be patient/tumor tailored with a shared plan about timing and type of imaging procedures to use to avoid safety issues. It is also stressed that patient-reported outcomes should receive greater attention, and that a multidisciplinary approach should be mandatory. Due to the clinical heterogeneity and relative lack of definitive evidence for NENs, personalization of diagnostic-therapeutic work-up is crucial.
Collapse
Affiliation(s)
- Mirco Bartolomei
- Azienda Ospedaliero-Universitaria di Ferrara, Presidio Ospedaliero Arcispedale Sant’Anna di Cona, 44124 Ferrara, Italy;
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Massimo Falconi
- Pancreas Surgical Unit, ENETS Center of Excellence, San Raffaele Hospital IRCCS, Vita Salute University, 20132 Milan, Italy;
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncologya and Neuroendocrine Tumors, European Institute of Oncology, 20132 Milan, Italy;
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties, IRCCS, Ospedale Policlinico San Martino, Università di Genova, 16132 Genova, Italy;
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, 80131 Naples, Italy;
| | - Giovanni Pappagallo
- School of Clinical Methodology IRCCS “Sacred Heart–Don Calabria” Hospital; 37024 Negrar di Valpolicella, Italy;
| | - Ettore Seregni
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20132 Milano, Italy;
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda Unità Sanitaria Locale-IRCCS of Reggio Emilia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
17
|
Hudson JM, Chung HTK, Chu W, Taggar A, Davis LE, Hallet J, Law CHL, Singh S, Myrehaug S. Stereotactic Ablative Radiotherapy for the Management of Liver Metastases from Neuroendocrine Neoplasms: A Preliminary Study. Neuroendocrinology 2022; 112:153-160. [PMID: 33530088 DOI: 10.1159/000514914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 01/21/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Liver metastases are common in patients with neuroendocrine neoplasms. The role of stereotactic ablative radiotherapy (SABR) is not well understood in this population. OBJECTIVE The objective of this study was to evaluate the safety and efficacy of SABR in treating well-differentiated neuroendocrine liver metastases (WD-NELM). METHODS A retrospective review of patients with WD-NELM treated with SABR was conducted between January 2015 and July 2019. Demographic, treatment, and clinical/radiographic follow-up data were abstracted. RECIST 1.1 criteria were applied to each individual target to evaluate the response to treatment. Local control (LC) and progression-free survival (PFS) were determined using the Kaplan-Meier methodology. Toxicity was reported according to the CTCAE v5.0. RESULTS Twenty-five patients with a total of 53 liver metastases treated with SABR were identified. Most patients (68%) had midgut tumors, were grade 2 (80%), and had high-volume intrahepatic and/or extrahepatic disease (76%). The median number of liver metastases treated was 2, with a median size of 2.5 cm. The median radiation dose delivered was 50 Gy/5 fractions. The median follow-up was 14 months; 24 of the 25 patients were alive at the time of analysis. The objective response rate was 32%, with improvement or stability in 96% of lesions treated. The median time to best response was 9 months. The 1-year LC and PFS were 92 and 44%, respectively. No grade 3/4 acute or late toxicity was identified. CONCLUSIONS Liver SABR is a safe and promising means of providing LC for WD-NELM. This treatment modality should be evaluated in selected patients in concert with strategies to manage systemic disease.
Collapse
Affiliation(s)
- John Monte Hudson
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hans Tse-Kan Chung
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - William Chu
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Amandeep Taggar
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Laura Ellen Davis
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Québec, Canada
| | - Julie Hallet
- Department of Surgical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Calvin How Lim Law
- Department of Surgical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Simron Singh
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Partouche E, Yeh R, Eche T, Rozenblum L, Carrere N, Guimbaud R, Dierickx LO, Rousseau H, Dercle L, Mokrane FZ. Updated Trends in Imaging Practices for Pancreatic Neuroendocrine Tumors (PNETs): A Systematic Review and Meta-Analysis to Pave the Way for Standardization in the New Era of Big Data and Artificial Intelligence. Front Oncol 2021; 11:628408. [PMID: 34336643 PMCID: PMC8316992 DOI: 10.3389/fonc.2021.628408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 06/25/2021] [Indexed: 01/03/2023] Open
Abstract
Purpose Medical imaging plays a central and decisive role in guiding the management of patients with pancreatic neuroendocrine tumors (PNETs). Our aim was to synthesize all recent literature of PNETs, enabling a comparison of all imaging practices. Methods based on a systematic review and meta-analysis approach, we collected; using MEDLINE, EMBASE, and Cochrane Library databases; all recent imaging-based studies, published from December 2014 to December 2019. Study quality assessment was performed by QUADAS-2 and MINORS tools. Results 161 studies consisting of 19852 patients were included. There were 63 ‘imaging’ studies evaluating the accuracy of medical imaging, and 98 ‘clinical’ studies using medical imaging as a tool for response assessment. A wide heterogeneity of practices was demonstrated: imaging modalities were: CT (57.1%, n=92), MR (42.9%, n=69), PET/CT (13.3%, n=31), and SPECT/CT (9.3%, n=15). International imaging guidelines were mentioned in 2.5% (n=4/161) of studies. In clinical studies, imaging protocol was not mentioned in 30.6% (n=30/98) of cases and only mentioned imaging modality without further information in 63.3% (n=62/98), as compared to imaging studies (1.6% (n=1/63) of (p<0.001)). QUADAS-2 and MINORS tools deciphered existing biases in the current literature. Conclusion We provide an overview of the updated current trends in use of medical imaging for diagnosis and response assessment in PNETs. The most commonly used imaging modalities are anatomical (CT and MRI), followed by PET/CT and SPECT/CT. Therefore, standardization and homogenization of PNETs imaging practices is needed to aggregate data and leverage a big data approach for Artificial Intelligence purposes.
Collapse
Affiliation(s)
- Ephraïm Partouche
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Randy Yeh
- Memorial Sloan Kettering Cancer Center, Molecular Imaging and Therapy Service., New York, NY, United States
| | - Thomas Eche
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Laura Rozenblum
- Sorbonne Université, Service de Médecine Nucléaire, AP-HP, Hôpital La Pitié-Salpêtrière, Paris, France
| | - Nicolas Carrere
- Surgery Department, Toulouse University Hospital, Toulouse, France
| | - Rosine Guimbaud
- Oncology Department, Toulouse University Hospital, Toulouse, France
| | | | - Hervé Rousseau
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Vagellos College of Physicians and Surgeons, New York, NY, United States
| | | |
Collapse
|
19
|
Koumarianou A, Kaltsas GA, Chatzellis E, Kyriakopoulos G, Kolomodi D, Alexandraki KI. Immunotherapeutics at the spearhead: current status in targeting neuroendocrine neoplasms. Endocrine 2021; 73:232-239. [PMID: 33544352 DOI: 10.1007/s12020-021-02639-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/18/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE Recent advances in the field of immunotherapy have significantly prolonged the survival of patients with aggressive carcinomas, but the role of immunotherapy in neuroendocrine neoplasms (NENs) remains to be elucidated. METHODS AND RESULTS We report a patient diagnosed with a well-differentiated grade 3 pancreatic NEN (pNEN) and type 3 liver metastases who received compassionate nivolumab as a fifth line treatment and achieved a durable partial response of more than 34 months. We have performed a systematic review to the literature on tumor microenvironment and potential biomarkers in the field of NEN including the tumor mutational burden, the tumor infiltrating lymphocytes, the programmed cell death ligand 1, and the mismatch repair system. The potential role of the immune system modulation together with a critical assessment of the recent phase II clinical studies in NEN including monotherapy with anti-PD-1/PD-L1 monoclonal antibodies, and combination therapies including anti-PD-1 along with anti-CTLA-4 monoclonal antibodies are also provided. CONCLUSION Immunotherapeutics are gaining a post in the field of NENs in cases progressing during the course of the disease, dictating urgently the identification of biomarkers that will enable selection of NEN patients who may benefit from this treatment.
Collapse
Affiliation(s)
- Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece.
| | - Gregory A Kaltsas
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
- 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Chatzellis
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | - Georgios Kyriakopoulos
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | - Denise Kolomodi
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | | |
Collapse
|
20
|
Wang R, Liu H, Liang P, Zhao H, Li L, Gao J. Radiomics analysis of CT imaging for differentiating gastric neuroendocrine carcinomas from gastric adenocarcinomas. Eur J Radiol 2021; 138:109662. [PMID: 33774440 DOI: 10.1016/j.ejrad.2021.109662] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/29/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE To develop and evaluate a CT-based radiomics nomogram for differentiating gastric neuroendocrine carcinomas (NECs) from gastric adenocarcinomas (ADCs). METHODS CT images of 63 patients with gastric NECs were collected retrospectively, and 63 patients with gastric ADCs were selected as the control group. Univariate analysis was used to identify the significant factors of clinical characteristics and CT findings for differentiating gastric NECs from ADCs. Radiomics analysis was applied to CT images of unenhanced, arterial phase and venous phase, respectively. A radiomics nomogram incorporating the radiomics signature and the subjective CT findings was developed and its diagnostic ability was evaluated. The diagnostic performances of CT findings model, radiomics signature and radiomics nomogram were compared using DeLong test. RESULTS The tumor margin and lymph node (LN) metastasis were independent predictors for differentiating gastric NECs from ADCs. The radiomics signature based on venous phase presented superior AUC of 0.798 [95 % confidence interval (CI), 0.657-0.938] in validation cohort. The nomogram incorporated the radiomics signature, tumor margin and LN metastasis showed AUCs of 0.821 (95 %CI: 0.725-0.895) in the primary cohort and 0.809 (95 %CI: 0.649-0.918) in the validation cohort. Moreover, the radiomics nomogram showed good discrimination and calibration. The diagnostic performance of CT findings model was significantly lower than that of radiomics nomogram (p = 0.001) and radiomics signature (p = 0.025). CONCLUSIONS Radiomics analysis exhibited good performance in differentiating gastric NECs from ADCs, and the radiomics nomogram may have significant clinical implications on preoperative detection of gastric malignant tumors.
Collapse
Affiliation(s)
- Rui Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Huan Liu
- Advanced Application Team, GE Healthcare, Shanghai, 201203, China
| | - Pan Liang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Huiping Zhao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liming Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
21
|
Baghdadi A, Ghadimi M, Mirpour S, Hazhirkarzar B, Motaghi M, Pawlik TM, Kamel IR. Imaging neuroendocrine tumors: Characterizing the spectrum of radiographic findings. Surg Oncol 2021; 37:101529. [PMID: 33549952 DOI: 10.1016/j.suronc.2021.101529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/25/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Neuroendocrine tumors (NET) are a group of neoplasms with neuroendocrine differentiation affecting a wide range of organs. Functional NETs present with symptoms due to the particular hormone produced. Functional NETs are usually small at diagnosis and therefore can be challenging to diagnose. In contrast, non-functioning NETs are generally larger and present with mass effect. Imaging plays an indispensable role in diagnosis, staging and management of patients with NETs. The optimal modality and technique for imaging of NETs depend on the location of primary and metastatic lesions. Regardless of the imaging modality, dynamic contrast-enhanced imaging is essential for evaluation of NETs. In general, CT scan is typically the primary imaging modality for evaluating NETs. MRI is used as a complementary modality, being superior to other modalities to assess liver metastasis. Nuclear medicine imaging is also widely used in NET assessment.
Collapse
Affiliation(s)
- Azarakhsh Baghdadi
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA
| | - Maryam Ghadimi
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA
| | - Sahar Mirpour
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA
| | - Bita Hazhirkarzar
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA
| | - Mina Motaghi
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Ihab R Kamel
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, 600 N Wolfe St, Room 143, Baltimore, MD, 21287, USA.
| |
Collapse
|
22
|
Xu J, Shen L, Zhou Z, Li J, Bai C, Chi Y, Li Z, Xu N, Li E, Liu T, Bai Y, Yuan Y, Li X, Wang X, Chen J, Ying J, Yu X, Qin S, Yuan X, Zhang T, Deng Y, Xiu D, Cheng Y, Tao M, Jia R, Wang W, Li J, Fan S, Peng M, Su W. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020; 21:1500-1512. [PMID: 32966811 DOI: 10.1016/s1470-2045(20)30496-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Therapeutic options for advanced neuroendocrine tumours (NETs) are limited. We investigated the efficacy and safety of surufatinib (HMPL-012, sulfatinib) in patients with extrapancreatic NETs. METHODS SANET-ep was a randomised, double-blind, placebo-controlled, phase 3 trial undertaken at 24 hospitals across China. Patients (aged 18 years or older) with unresectable or metastatic, well differentiated, extrapancreatic NETs, with an Eastern Cooperative Oncology Group performance status of 0 or 1, and progression on no more than two types of previous systemic regimens were enrolled. Patients were centrally randomly assigned (2:1) using stratified block randomisation (block size 3) via an interactive web response system to receive oral surufatinib at 300 mg per day or matching placebo. Randomisation was stratified by tumour origin, pathological grade, and previous treatment. Patients, investigators, research staff and the sponsor study team were masked to treatment allocation. Crossover to the surufatinib group was allowed for patients in the placebo group at disease progression. The primary endpoint was investigator-assessed progression-free survival, which was analysed in the intention-to-treat population. A preplanned interim analysis was done at 70% of predicted progression-free survival events. This study was registered with ClinicalTrials.gov, NCT02588170. Follow-up is ongoing. FINDINGS Between Dec 9, 2015, and March 31, 2019, 198 patients were randomly assigned to surufatinib (n=129) or placebo (n=69). Median follow-up was 13·8 months (95% CI 11·1-16·7) in the surufatinib group and 16·6 months (9·2-not calculable) in the placebo group. Investigator-assessed median progression-free survival was 9·2 months (95% CI 7·4-11·1) in the surufatinib group versus 3·8 months (3·7-5·7) in the placebo group (hazard ratio 0·33; 95% CI 0·22-0·50; p<0·0001). As the trial met the predefined criteria for early discontinuation of the study at the interim analysis, the study was terminated early, as recommended by the independent data monitoring committee. The most common treatment-related adverse events of grade 3 or worse were hypertension (47 [36%] of 129 patients in the surufatinib group vs nine [13%] of 68 patients in the placebo group) and proteinuria (25 [19%] vs zero). Treatment-related serious adverse events were reported in 32 (25%) of 129 patients in the surufatinib group and nine (13%) of 68 patients in the placebo group. Treatment-related deaths occurred in three patients in the surufatinib group (disseminated intravascular coagulation and hepatic encephalopathy, liver injury, and death with unknown reason) and one patient in the placebo group (cachexia and respiratory failure). INTERPRETATION Progression-free survival was significantly longer in patients given surufatinib compared with patients given placebo, and surufatinib has a favourable benefit-to-risk profile in patients with progressive, advanced, well differentiated extrapancreatic NETs. Our results suggest that surufatinib might be a new treatment option for this population. FUNDING Hutchison MediPharma.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jie Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Beijing, China
| | - Yihebali Chi
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiping Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Yuan
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiuwen Wang
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Jia Chen
- Department of Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Jieer Ying
- Department of Abdominal Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shukui Qin
- People's Liberation Army Cancer Center of Nanjing Jinling Hospital, Nanjing, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases l, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ru Jia
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Wang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Li
- Department of Clinical Development and Regulatory Affairs, Hutchison MediPharma, Shanghai, China
| | - Songhua Fan
- Department of Clinical Development and Regulatory Affairs, Hutchison MediPharma, Shanghai, China
| | - Mengye Peng
- Department of Clinical Development and Regulatory Affairs, Hutchison MediPharma, Shanghai, China
| | - Weiguo Su
- Department of Clinical Development and Regulatory Affairs, Hutchison MediPharma, Shanghai, China
| |
Collapse
|
23
|
Werner RA, Hänscheid H, Leal JP, Javadi MS, Higuchi T, Lodge MA, Buck AK, Pomper MG, Lapa C, Rowe SP. Impact of Tumor Burden on Quantitative [ 68Ga] DOTATOC Biodistribution. Mol Imaging Biol 2020; 21:790-798. [PMID: 30406512 DOI: 10.1007/s11307-018-1293-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE As has been previously reported, the somatostatin receptor (SSTR) imaging agent [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotate ([68Ga]DOTATATE) demonstrates lower uptake in normal organs in patients with a high neuroendocrine tumor (NET) burden. Given the higher SSTR affinity of [68Ga] DOTATATE, we aimed to quantitatively investigate the biodistribution of [68Ga]-labeled 1,4,7,10-tetraazacyclododecane-N,N',N″,N‴-tetraacetic acid-d-Phe(1)-Tyr(3)-octreotide ([68Ga]DOTATOC) to determine a potential correlation between uptake in normal organs and NET burden. PROCEDURES Of the 44 included patients, 36/44 (82 %) patients demonstrated suspicious radiotracer uptake on [68Ga] DOTATOC positron emission tomography (PET)/X-ray computed tomography (CT). Volumes of interest (VOIs) were defined for tumor lesions and normal organs (spleen, liver, kidneys, adrenals). Mean body weight corrected standardized uptake value (SUVmean) for normal organs was assessed and was used to calculate the corresponding mean specific activity uptake (Upt: fraction of injected activity per kg of tissue). For the entire tumor burden, SUVmean, maximum standardized uptake value (SUVmax), and the total mass (TBM) was calculated and the decay corrected tumor fractional uptake (TBU) was assessed. A Spearman's rank correlation coefficient was used to determine the correlations between normal organ uptake and tumor burden. RESULTS The median SUVmean was 18.7 for the spleen (kidneys, 9.2; adrenals, 6.8; liver, 5.6). For tumor burden, the median values were SUVmean 6.9, SUVmax 35.5, TBM 42.6 g, and TBU 1.2 %. With increasing volume of distribution, represented by lean body mass and body surface area (BSA), Upt decreased in kidneys, liver, and adrenal glands and SUVmean increased in the spleen. Correlation improved only for both kidneys and adrenals when the influence of the tumor uptake on the activity available for organ uptake was taken into account by the factor 1/(1-TBU). TBU was neither predictive for SUVmean nor for Upt in any of the organs. The distribution of organ Upt vs. BSA/(1-TBU) were not different for patients with minor TBU (<3 %) vs. higher TBU (>7 %), indicating that the correlations observed in the present study are explainable by the body size effect. High tumor mass and uptake mitigated against G1 NET. CONCLUSIONS There is no significant impact on normal organ biodistribution with increasing tumor burden on [68Ga] DOTATOC PET/CT. Potential implications include increased normal organ dose with [177Lu-DOTA]0-D-Phe1-Tyr3-Octreotide and decreased absolute lesion detection with [68Ga] DOTATOC in high NET burden.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany.,The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Heribert Hänscheid
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jeffrey P Leal
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Mehrbod S Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin A Lodge
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.,European Neuroendocrine Tumor Society (ENETS) Center of Excellence, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, 601 N. Caroline St, Baltimore, MD, 21287, USA. .,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Rozenblum L, Mokrane FZ, Yeh R, Sinigaglia M, Besson FL, Seban RD, Zadro C, Dierickx L, Chougnet CN, Partouche E, Revel-Mouroz P, Zhao B, Otal P, Schwartz LH, Dercle L. Imaging-guided precision medicine in non-resectable gastro-entero-pancreatic neuroendocrine tumors: A step-by-step approach. Eur J Radiol 2020; 122:108743. [DOI: 10.1016/j.ejrad.2019.108743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/11/2019] [Indexed: 12/11/2022]
|
25
|
Early Prediction of Treatment Response of Neuroendocrine Hepatic Metastases after Peptide Receptor Radionuclide Therapy with 90Y-DOTATOC Using Diffusion Weighted and Dynamic Contrast-Enhanced MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:1517208. [PMID: 31787860 PMCID: PMC6877975 DOI: 10.1155/2019/1517208] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/11/2019] [Indexed: 01/20/2023]
Abstract
The purpose of this study was to determine if parameters derived from diffusion-weighted (DW-) and dynamic contrast-enhanced (DCE-) magnetic resonance imaging (MRI) can help to assess early response to peptide receptor radionuclide therapy (PRRT) with 90Y-DOTATOC in neuroendocrine hepatic metastases (NET-HM). Twenty patients (10 male; 10 female; mean age: 59.2 years) with NET-HM were prospectively enrolled in this single-center imaging study. DW-MRI and DCE-MRI studies were performed just before and 48 hours after therapy with 90Y-DOTATOC. Abdominal SPECT/CT was performed 24 hours after therapy. This MRI imaging and therapy session was repeated after a mean interval of 10 weeks. Up to four lesions per patient were evaluated. Response to therapy was evaluated using metastasis sizes at the first and second therapy session as standard for comparison (regressive, stable, and progressive). DW-MRI analysis included the apparent diffusion coefficient (ADC) and parameters related to intravoxel incoherent motion (IVIM), namely, diffusion (D), perfusion fraction (f) and pseudo-diffusion (D ∗ ). DCE-MRI analysis comprised Ktrans, v e and k ep. For statistical analysis of group differences, one-way analysis of variance (ANOVA) and appropriate post hoc testing was performed. A total of 51 lesions were evaluated. Seven of 51 lesions (14%) showed size progression, 18/51 (35%) regression, and 26/51 (51%) remained stable. The lesion-to-spleen uptake ratio in SPECT showed a decrease between the two treatment sessions that was significantly stronger in regressive lesions compared with stable (p = 0.013) and progressive lesions (p = 0.021). ANOVA showed significant differences in mean ADC after 48 h (p = 0.026), with higher ADC values for regressive lesions. Regarding IVIM, highest values for D at baseline were seen in regressive lesions (p = 0.023). In DCE-MRI, a statistically significant increase in v e after 10 weeks (p = 0.046) was found in regressive lesions. No differences were observed for the transfer constants Ktrans and k ep. Diffusion restriction quantified as ADC was able to differentiate regressive from progressive NET-HMs as early as 48 hours after PRRT. DW-MRI therefore may complement scintigraphy/SPECT for early assessment of response to PRRT. Assessment of perfusion parameters using IVIM and DCE-MRI did not show an additional benefit.
Collapse
|
26
|
Taïeb D, Jha A, Treglia G, Pacak K. Molecular imaging and radionuclide therapy of pheochromocytoma and paraganglioma in the era of genomic characterization of disease subgroups. Endocr Relat Cancer 2019; 26:R627-R652. [PMID: 31561209 PMCID: PMC7002202 DOI: 10.1530/erc-19-0165] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
In recent years, advancement in genetics has profoundly helped to gain a more comprehensive molecular, pathogenic, and prognostic picture of pheochromocytomas and paragangliomas (PPGLs). Newly discovered molecular targets, particularly those that target cell membranes or signaling pathways have helped move nuclear medicine in the forefront of PPGL precision medicine. This is mainly based on the introduction and increasing experience of various PET radiopharmaceuticals across PPGL genotypes quickly followed by implementation of novel radiotherapies and revised imaging algorithms. Particularly, 68Ga-labeled-SSAs have shown excellent results in the diagnosis and staging of PPGLs and in selecting patients for PRRT as a potential alternative to 123/131I-MIBG theranostics. PRRT using 90Y/177Lu-DOTA-SSAs has shown promise for treatment of PPGLs with improvement of clinical symptoms and/or disease control. However, more well-designed prospective studies are required to confirm these findings, in order to fully exploit PRRT's antitumoral properties to obtain the final FDA approval. Such an approval has recently been obtained for high-specific-activity 131I-MIBG for inoperable/metastatic PPGL. The increasing experience and encouraging preliminary results of these radiotherapeutic approaches in PPGLs now raises an important question of how to further integrate them into PPGL management (e.g. monotherapy or in combination with other systemic therapies), carefully taking into account the PPGLs locations, genotypes, and growth rate. Thus, targeted radionuclide therapy (TRT) should preferably be performed at specialized centers with an experienced interdisciplinary team. Future perspectives include the introduction of dosimetry and biomarkers for therapeutic responses for more individualized treatment plans, α-emitting isotopes, and the combination of TRT with other systemic therapies.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio Treglia
- Clinic of Nuclear Medicine and PET/CT Center, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- Health Technology Assessment Unit, General Directorate, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Evaluating radiological response in pancreatic neuroendocrine tumours treated with sunitinib: comparison of Choi versus RECIST criteria (CRIPNET_ GETNE1504 study). Br J Cancer 2019; 121:537-544. [PMID: 31477779 PMCID: PMC6889276 DOI: 10.1038/s41416-019-0558-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/09/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The purpose of our study was to analyse the usefulness of Choi criteria versus RECIST in patients with pancreatic neuroendocrine tumours (PanNETs) treated with sunitinib. METHOD A multicentre, prospective study was conducted in 10 Spanish centres. Computed tomographies, at least every 6 months, were centrally evaluated until tumour progression. RESULTS One hundred and seven patients were included. Median progression-free survival (PFS) by RECIST and Choi were 11.42 (95% confidence interval [CI], 9.7-15.9) and 15.8 months (95% CI, 13.9-25.7). PFS by Choi (Kendall's τ = 0.72) exhibited greater correlation with overall survival (OS) than PFS by RECIST (Kendall's τ = 0.43). RECIST incorrectly estimated prognosis in 49.6%. Partial response rate increased from 12.8% to 47.4% with Choi criteria. Twenty-four percent of patients with progressive disease according to Choi had stable disease as per RECIST, overestimating treatment effect. Choi criteria predicted PFS/OS. Changes in attenuation occurred early and accounted for 21% of the variations in tumour volume. Attenuation and tumour growth rate (TGR) were associated with improved survival. CONCLUSION Choi criteria were able to capture sunitinib's activity in a clinically significant manner better than RECIST; their implementation in standard clinical practice shall be strongly considered in PanNET patients treated with this drug.
Collapse
|
28
|
Carmona-Bayonas A, Jiménez-Fonseca P, Lamarca Á, Barriuso J, Castaño Á, Benavent M, Alonso V, Riesco-Martínez MDC, Alonso-Gordoa T, Custodio A, Sánchez Cánovas M, Hernando Cubero J, López C, Lacasta A, Fernández Montes A, Marazuela M, Crespo G, Escudero P, Diaz JÁ, Feliciangeli E, Gallego J, Llanos M, Segura Á, Vilardell F, Percovich JC, Grande E, Capdevila J, Valle JW, García-Carbonero R. Prediction of Progression-Free Survival in Patients With Advanced, Well-Differentiated, Neuroendocrine Tumors Being Treated With a Somatostatin Analog: The GETNE-TRASGU Study. J Clin Oncol 2019; 37:2571-2580. [PMID: 31390276 PMCID: PMC6768612 DOI: 10.1200/jco.19.00980] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Somatostatin analogs (SSAs) are recommended for the first-line treatment of most patients with well-differentiated, gastroenteropancreatic (GEP) neuroendocrine tumors; however, benefit from treatment is heterogeneous. The aim of the current study was to develop and validate a progression-free survival (PFS) prediction model in SSA-treated patients. PATIENTS AND METHODS We extracted data from the Spanish Group of Neuroendocrine and Endocrine Tumors Registry (R-GETNE). Patient eligibility criteria included GEP primary, Ki-67 of 20% or less, and first-line SSA monotherapy for advanced disease. An accelerated failure time model was developed to predict PFS, which was represented as a nomogram and an online calculator. The nomogram was externally validated in an independent series of consecutive eligible patients (The Christie NHS Foundation Trust, Manchester, United Kingdom). RESULTS We recruited 535 patients (R-GETNE, n = 438; Manchester, n = 97). Median PFS and overall survival in the derivation cohort were 28.7 (95% CI, 23.8 to 31.1) and 85.9 months (95% CI, 71.5 to 96.7 months), respectively. Nine covariates significantly associated with PFS were primary tumor location, Ki-67 percentage, neutrophil-to-lymphocyte ratio, alkaline phosphatase, extent of liver involvement, presence of bone and peritoneal metastases, documented progression status, and the presence of symptoms when initiating SSA. The GETNE-TRASGU (Treated With Analog of Somatostatin in Gastroenteropancreatic and Unknown Primary NETs) model demonstrated suitable calibration, as well as fair discrimination ability with a C-index value of 0.714 (95% CI, 0.680 to 0.747) and 0.732 (95% CI, 0.658 to 0.806) in the derivation and validation series, respectively. CONCLUSION The GETNE-TRASGU evidence-based prognostic tool stratifies patients with GEP neuroendocrine tumors receiving SSA treatment according to their estimated PFS. This nomogram may be useful when stratifying patients with neuroendocrine tumors in future trials. Furthermore, it could be a valuable tool for making treatment decisions in daily clinical practice.
Collapse
Affiliation(s)
- Alberto Carmona-Bayonas
- Hospital Universitario Morales Meseguer, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | | | - Ángela Lamarca
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Jorge Barriuso
- The Christie NHS Foundation Trust, Manchester, United Kingdom.,University of Manchester, Manchester, United Kingdom
| | - Ángel Castaño
- Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Marta Benavent
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | | | | | | | - Ana Custodio
- Hospital Universitario La Paz, Centro de Investigación Biomédica en Red Cáncer, CB16/12/00398, Madrid, Spain
| | - Manuel Sánchez Cánovas
- Hospital Universitario Morales Meseguer, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Jorge Hernando Cubero
- Hospital Universitario Vall d'Hebron, Vall Hebron Institute of Oncology, Autonomous University of Barcelona, Barcelona, Spain
| | - Carlos López
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | - Pilar Escudero
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | | | | | | | - Marta Llanos
- Hospital Universitario de Canarias, Universidad de La Laguna, Tenerife, Spain
| | | | | | | | | | - Jaume Capdevila
- Hospital Universitario Vall d'Hebron, Vall Hebron Institute of Oncology, Autonomous University of Barcelona, Barcelona, Spain
| | - Juan W Valle
- The Christie NHS Foundation Trust, Manchester, United Kingdom.,University of Manchester, Manchester, United Kingdom
| | - Rocío García-Carbonero
- Hospital Universitario Doce de Octubre, Universidad Complutense de Madrid, Centro Nacional de Investigaciones Oncológicas, Centro de Investigación Biomédica en Red Cáncer, Madrid, Spain
| |
Collapse
|
29
|
Werner RA, Thackeray JT, Pomper MG, Bengel FM, Gorin MA, Derlin T, Rowe SP. Recent Updates on Molecular Imaging Reporting and Data Systems (MI-RADS) for Theranostic Radiotracers-Navigating Pitfalls of SSTR- and PSMA-Targeted PET/CT. J Clin Med 2019; 8:E1060. [PMID: 31331016 PMCID: PMC6678732 DOI: 10.3390/jcm8071060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The theranostic concept represents a paradigmatic example of personalized treatment. It is based on the use of radiolabeled compounds which can be applied for both diagnostic molecular imaging and subsequent treatment, using different radionuclides for labelling. Clinically relevant examples include somatostatin receptor (SSTR)-targeted imaging and therapy for the treatment of neuroendocrine tumors (NET), as well as prostate-specific membrane antigen (PSMA)-targeted imaging and therapy for the treatment of prostate cancer (PC). As such, both classes of radiotracers can be used to triage patients for theranostic endoradiotherapy using positron emission tomography (PET). While interpreting PSMA- or SSTR-targeted PET/computed tomography scans, the reader has to navigate certain pitfalls, including (I.) varying normal biodistribution between different PSMA- and SSTR-targeting PET radiotracers, (II.) varying radiotracer uptake in numerous kinds of both benign and malignant lesions, and (III.) resulting false-positive and false-negative findings. Thus, two novel reporting and data system (RADS) classifications for PSMA- and SSTR-targeted PET imaging (PSMA- and SSTR-RADS) have been recently introduced under the umbrella term molecular imaging reporting and data systems (MI-RADS). Notably, PSMA- and SSTR-RADS are structured in a reciprocal fashion, i.e., if the reader is familiar with one system, the other system can readily be applied. Learning objectives of the present case-based review are as follows: (I.) the theranostic concept for the treatment of NET and PC will be briefly introduced, (II.) the most common pitfalls on PSMA- and SSTR-targeted PET/CT will be identified, (III.) the novel framework system for theranostic radiotracers (MI-RADS) will be explained, applied to complex clinical cases and recent studies in the field will be highlighted. Finally, current treatment strategies based on MI-RADS will be proposed, which will demonstrate how such a generalizable framework system truly paves the way for clinically meaningful molecular imaging-guided treatment of either PC or NET. Thus, beyond an introduction of MI-RADS, the present review aims to provide an update of recently published studies which have further validated the concept of structured reporting systems in the field of theranostics.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Martin G Pomper
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Michael A Gorin
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Steven P Rowe
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA.
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
30
|
Panda A, Garg I, Johnson GB, Truty MJ, Halfdanarson TR, Goenka AH. Molecular radionuclide imaging of pancreatic neoplasms. Lancet Gastroenterol Hepatol 2019; 4:559-570. [DOI: 10.1016/s2468-1253(19)30081-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/26/2019] [Accepted: 03/02/2019] [Indexed: 02/07/2023]
|
31
|
Werner RA, Ilhan H, Lehner S, Papp L, Zsótér N, Schatka I, Muegge DO, Javadi MS, Higuchi T, Buck AK, Bartenstein P, Bengel F, Essler M, Lapa C, Bundschuh RA. Pre-therapy Somatostatin Receptor-Based Heterogeneity Predicts Overall Survival in Pancreatic Neuroendocrine Tumor Patients Undergoing Peptide Receptor Radionuclide Therapy. Mol Imaging Biol 2019; 21:582-590. [PMID: 30014345 DOI: 10.1007/s11307-018-1252-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Early identification of aggressive disease could improve decision support in pancreatic neuroendocrine tumor (pNET) patients prior to peptide receptor radionuclide therapy (PRRT). The prognostic value of intratumoral textural features (TF) determined by baseline somatostatin receptor (SSTR)-positron emission tomography (PET) before PRRT was analyzed. PROCEDURES Thirty-one patients with G1/G2 pNET were enrolled (G2, n = 23/31). Prior to PRRT with [177Lu]DOTATATE (mean, 3.6 cycles), baseline SSTR-PET computed tomography was performed. By segmentation of 162 (median per patient, 5) metastases, intratumoral TF were computed. The impact of conventional PET parameters (SUVmean/max), imaging-based TF, and clinical parameters (Ki67, CgA) for prediction of both progression-free survival (PFS) and overall survival (OS) after PRRT were evaluated. RESULTS Within a median follow-up of 3.7 years, tumor progression was detected in 21 patients (median, 1.5 years) and 13/31 deceased (median, 1.9 years). In ROC analysis, the TF entropy, reflecting derangement on a voxel-by-voxel level, demonstrated predictive capability for OS (cutoff = 6.7, AUC = 0.71, p = 0.02). Of note, increasing entropy could predict a longer survival (> 6.7, OS = 2.5 years, 17/31), whereas less voxel-based derangement portended inferior outcome (< 6.7, OS = 1.9 years, 14/31). These findings were supported in a G2 subanalysis (> 6.9, OS = 2.8 years, 9/23 vs. < 6.9, OS = 1.9 years, 14/23). Kaplan-Meier analysis revealed a significant distinction between high- and low-risk groups using entropy (n = 31, p < 0.05). For those patients below the ROC-derived threshold, the relative risk of death after PRRT was 2.73 (n = 31, p = 0.04). Ki67 was negatively associated with PFS (p = 0.002); however, SUVmean/max failed in prognostication (n.s.). CONCLUSIONS In contrast to conventional PET parameters, assessment of intratumoral heterogeneity demonstrated superior prognostic performance in pNET patients undergoing PRRT. This novel PET-based strategy of outcome prediction prior to PRRT might be useful for patient risk stratification.
Collapse
Affiliation(s)
- Rudolf A Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Ambulatory Healthcare Center Dr. Neumaier & Colleagues, Radiology, Nuclear Medicine, Radiation Therapy, Regensburg, Germany
| | - László Papp
- Department of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Imke Schatka
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dirk O Muegge
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Mehrbod S Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Department of Bio Medical Imaging, National Cardiovascular and Cerebral Research Center, Suita, Japan
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Frank Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Markus Essler
- Department of Nuclear Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany.
| |
Collapse
|
32
|
Therapy With 177Lu-DOTATATE: Clinical Implementation and Impact on Care of Patients With Neuroendocrine Tumors. AJR Am J Roentgenol 2019; 213:309-317. [PMID: 31039017 DOI: 10.2214/ajr.19.21123] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE. The purpose of this article is to enhance knowledge of the clinical implementation of peptide receptor radionuclide therapy (PRRT) and its impact on care of patients with neuroendocrine tumors. CONCLUSION. Most well differentiated and some moderately and poorly differentiated neuroendocrine tumors express large numbers of somatostatin receptors on their cell surfaces. PRRT targets these cells with 177Lu-DOTATATE, which is a medium-energy beta emitter. Since this agent received U.S. Food and Drug Administration approval in 2018, tremendous effort has been exerted at institutions throughout the United States toward proper implementation of this promising therapy. This review summarizes clinical implementation of PRRT and its impact on patient care.
Collapse
|
33
|
Peptide Receptor Radionuclide Therapy for Patients With Advanced Lung Carcinoids. Clin Lung Cancer 2019; 20:e376-e392. [PMID: 30910575 DOI: 10.1016/j.cllc.2019.02.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/01/2019] [Accepted: 02/16/2019] [Indexed: 12/13/2022]
Abstract
Neuroendocrine neoplasms (NEN) are a family of malignancies of diverse origin, including the lung, gastrointestinal tract, and pancreas. Lung NEN include well differentiated neuroendocrine tumors (NET) classified as typical carcinoids or atypical carcinoids, and poorly differentiated neuroendocrine carcinomas classified as small-cell lung carcinoma or large-cell neuroendocrine carcinoma. According to a recent analysis of a large, population-based registry, approximately one-third of all patients with lung typical/atypical carcinoids have distant metastases at diagnosis, and median survival for these patients is 24 months. At present, only 1 therapy is approved by the US Food and Drug Administration (FDA) for patients with advanced lung typical/atypical carcinoids, everolimus, indicating a clear need for more treatment options in this patient population. Although not yet supported by results from randomized prospective trials, somatostatin analogues are considered an acceptable treatment option for patients with lung typical/atypical carcinoids expressing somatostatin receptors. Peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE was recently approved by the FDA for the treatment of gastroenteropancreatic NET; however, the role of PRRT in patients with lung typical/atypical carcinoids remains unclear, because they were not included in the pivotal NETTER-1 (Neuroendocrine Tumors Therapy) trial. Herein we provide a comprehensive review of the available clinical evidence for efficacy and safety of PRRT in patients with lung typical/atypical carcinoids. On the basis of the preliminary evidence of efficacy and the consistent safety profile in this patient group, we propose that experienced multidisciplinary NET teams may consider PRRT alongside everolimus as an option for patients with advanced somatostatin receptor-positive lung typical/atypical carcinoids whose disease is progressing during first-line treatment with somatostatin analogues.
Collapse
|
34
|
Hendifar AE, Dhall D, Strosberg JR. The Evolving Treatment Algorithm for Advanced Neuroendocrine Neoplasms: Diversity and Commonalities Across Tumor Types. Oncologist 2019; 24:54-61. [PMID: 30104288 PMCID: PMC6324634 DOI: 10.1634/theoncologist.2018-0187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/05/2018] [Indexed: 12/14/2022] Open
Abstract
Neuroendocrine neoplasms (NEN) most commonly arise in the gastroenteropancreatic system and lungs. The incidence of NEN is increasing globally, with improved diagnostic techniques identifying patients with early-stage disease. The number of approved therapies for the treatment of advanced disease has grown substantially in the past decade. The treatment algorithm for advanced NEN is evolving from one that is directed by primary site-specific classification to one that is directed by biologic classification, as evidenced by overlapping systemic treatments across the primary tumor sites. Commonalities in biologic characteristics across primary sites include functional status, differentiation status, grade, level of somatostatin receptor expression, and genetic alterations. In this review, we discuss current clinical evidence and available therapies for the treatment of advanced NEN and highlight the need for prospective trials in patients with well-differentiated, high-grade NEN. IMPLICATIONS FOR PRACTICE: This review raises awareness of the evolution of the treatment algorithm for advanced neuroendocrine neoplasms (NEN) from one that is directed by primary tumor site-specific classification to one that is directed by biologic classification. In addition, this review promotes understanding of the new pathologic category of well-differentiated G3 pancreatic neuroendocrine tumors and highlights the need for prospective trials in this patient population, for whom there is currently no standard of care. This review further provides a conceptual treatment schematic that categorizes the recommendations for systemic treatments for advanced disease by biologic classification, including the new and established categories of NEN.
Collapse
Affiliation(s)
- Andrew E Hendifar
- Department of Medicine and Pathology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Deepti Dhall
- Department of Medicine and Pathology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | |
Collapse
|
35
|
Werner RA, Weich A, Kircher M, Solnes LB, Javadi MS, Higuchi T, Buck AK, Pomper MG, Rowe SP, Lapa C. The theranostic promise for Neuroendocrine Tumors in the late 2010s - Where do we stand, where do we go? Theranostics 2018; 8:6088-6100. [PMID: 30613284 PMCID: PMC6299695 DOI: 10.7150/thno.30357] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
More than 25 years after the first peptide receptor radionuclide therapy (PRRT), the concept of somatostatin receptor (SSTR)-directed imaging and therapy for neuroendocrine tumors (NET) is seeing rapidly increasing use. To maximize the full potential of its theranostic promise, efforts in recent years have expanded recommendations in current guidelines and included the evaluation of novel theranostic radiotracers for imaging and treatment of NET. Moreover, the introduction of standardized reporting framework systems may harmonize PET reading, address pitfalls in interpreting SSTR-PET/CT scans and guide the treating physician in selecting PRRT candidates. Notably, the concept of PRRT has also been applied beyond oncology, e.g. for treatment of inflammatory conditions like sarcoidosis. Future perspectives may include the efficacy evaluation of PRRT compared to other common treatment options for NET, novel strategies for closer monitoring of potential side effects, the introduction of novel radiotracers with beneficial pharmacodynamic and kinetic properties or the use of supervised machine learning approaches for outcome prediction. This article reviews how the SSTR-directed theranostic concept is currently applied and also reflects on recent developments that hold promise for the future of theranostics in this context.
Collapse
Affiliation(s)
- Rudolf A. Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Alexander Weich
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Germany
| | - Malte Kircher
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Lilja B. Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mehrbod S. Javadi
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- Department of Bio Medical Imaging, National Cardiovascular and Cerebral Research Center, Suita, Japan
| | - Andreas K. Buck
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Constantin Lapa
- Department of Nuclear Medicine/Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence (CoE), NET Zentrum, University Hospital Würzburg, Germany
| |
Collapse
|
36
|
Merino-Casabiel X, Aller J, Arbizu J, García-Figueiras R, González C, Grande E, Jiménez-Fonseca P, Sevilla MI, Capdevila J. Consensus document on the progression and treatment response criteria in gastroenteropancreatic neuroendocrine tumors. Clin Transl Oncol 2018; 20:1522-1528. [PMID: 29766455 PMCID: PMC6223716 DOI: 10.1007/s12094-018-1881-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Gastroenteropancreatic neuroendocrine tumors are a heterogeneous group of low incidence neoplasms characterized by a low proliferative activity and slow growth. Their response to targeted therapies is heterogeneous and often does not lead to tumor shrinkage. Thus, evaluation of the therapeutic response should differ from other kind of tumors. METHODS To answer relevant questions about which techniques are best in the assessment of progression or treatment response a RAND/UCLA-based consensus process was implemented. Relevant clinical questions were listed followed by a systematic search of the literature. The expert panel answered all questions with recommendations, combining available evidence and expert opinion. Recommendations were validated through a questionnaire and a participatory meeting. RESULTS Expert recommendations regarding imaging tools for tumor assessment and evaluation of progression were agreed upon. Available imaging techniques were reviewed and recommendations for best patient monitoring practice and the best way to evaluate treatment response were formulated.
Collapse
Affiliation(s)
- X Merino-Casabiel
- Radiology Department, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - J Aller
- Endocrinology Department, Hospital Universitario Puerta de Hierro de Majadahonda, Madrid, Spain
| | - J Arbizu
- Nuclear Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - R García-Figueiras
- Radiology Department, Complexo Hospitalario Universitario de Santiago de Compostela, A Coruña, Spain
| | - C González
- Radiology Department, Hospital Universitario Puerta de Hierro de Majadahonda, Madrid, Spain
| | - E Grande
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - P Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - M I Sevilla
- Medical Oncology Department, Investigación Clínica y Traslacional en Cáncer, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Hospital Universitario Regional y Virgen de la Victoria de Málaga, Malaga, Spain
| | - J Capdevila
- Medical Oncology Department and Gastrointestinal and Endocrine Tumor Unit, Hospital Universitario Vall d'Hebron, Vall d'Hebron Institute of Oncology (VHIO), Pg Vall d'Hebron, 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
37
|
Tan TH, Boey CY, Lee BN. Impact of 68Ga-DOTA-Peptide PET/CT on the Management of Gastrointestinal Neuroendocrine Tumour (GI-NET): Malaysian National Referral Centre Experience. Nucl Med Mol Imaging 2018; 52:119-124. [PMID: 29662560 DOI: 10.1007/s13139-017-0496-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 08/23/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022] Open
Abstract
Purpose The National Cancer Institute is the only referral centre in Malaysia that provides 68Ga-DOTA-peptide imaging. The purpose of this study is to determine the impact of 68Ga-DOTA-peptide PET/CT on the management of gastrointestinal neuroendocrine tumours (GI-NET). Materials and Methods A cross-sectional study was performed to review the impact of 68Ga-DOTA-peptide (68Ga-DOTATATE or 68Ga-DOTATOC) PET/CT on patients with biopsy-proven GI-NET between January 2011 and December 2015. Suspected NET was excluded. Demographic data, tumoral characteristics, change of disease stage, pre-PET intended management and post-PET management were evaluated. Results Over a 5-year period, 82 studies of 68Ga-DOTA-peptide PET/CT were performed on 44 GI-NET patients. The most common primary site was the rectum (50.0%) followed by the small bowel, stomach and colon. Using WHO 2010 grading, 40.9% of patients had low-grade (G1) tumour, 22.7% intermediate (G2) and 4.5% high (G3). Of ten patients scheduled for pre-operative staging, 68Ga-DOTA-peptide PET/CT only led to therapeutic change in three patients. Furthermore, false-negative results of 68Ga-DOTA-peptide PET/CT were reported in one patient after surgical confirmation. However, therapeutic changes were seen in 20/36 patients (55.6%) scheduled for post-surgical restaging or assessment of somatostatin analogue (SSA) eligibility. When 68Ga-DOTA-peptide PET/CT was used for monitoring disease progress during systemic treatment (sandostatin, chemotherapy, everolimus and PRRT) in metastatic disease, impact on management modification was seen in 19/36 patients (52.8%), of which 84.2% had inter-modality change (switch to everolimus, chemotherapy or PRRT) and 15.8% had intra-modality change (increased SSA dosage). Conclusions 68Ga-DOTA-peptide PET/CT has a significant impact on management decisions in GI-NET patients as it can provide additional information on occult metastasis/equivocal lesions and supply the clinician an opportunity to select patients for targeted therapy.
Collapse
Affiliation(s)
- Teik Hin Tan
- Nuclear Medicine Centre, Sunway Medical Centre, No 5, Jalan Lagoon Selatan, 74500 Bandar Sunway, Selangor Malaysia
| | - Ching Yeen Boey
- 2Department of Nuclear Medicine, National Cancer Institute, No 4, Jalan P7, Presint 7, 62250 Putrajaya, Malaysia
| | - Boon Nang Lee
- 2Department of Nuclear Medicine, National Cancer Institute, No 4, Jalan P7, Presint 7, 62250 Putrajaya, Malaysia
| |
Collapse
|
38
|
Tumor Cystic Necrosis Following Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumors. Clin Nucl Med 2018; 43:186-187. [PMID: 29356741 DOI: 10.1097/rlu.0000000000001970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The response assessment to peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors is complex. We present the case of a 49-year-old man with metastatic rectal neuroendocrine tumor whose clinical symptoms and response evaluation criteria in solid tumors suggested progressive disease following PRRT. However, Ga-DOTA-(Tyr3)-octreotate PET/CT showed a partial scintigraphic response with absence of F-FDG PET/CT uptake consistent with tumor cystic necrosis. Long-term follow-up confirmed ongoing tumor response to treatment. Utilizing all modalities of response assessment seems to be important in correctly judging the benefit from PRRT and will need to be incorporated when developing response assessment tools.
Collapse
|
39
|
Computed diffusion weighted imaging (cDWI) and voxelwise-computed diffusion weighted imaging (vcDWI) for oncologic liver imaging: A pilot study. Eur J Radiol Open 2018; 5:108-113. [PMID: 30101156 PMCID: PMC6084526 DOI: 10.1016/j.ejro.2018.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/21/2018] [Accepted: 07/21/2018] [Indexed: 12/04/2022] Open
Abstract
Objective Aim of the study was to evaluate the influence of the selection of measured b-values on the precision of cDWI in the upper abdomen as well as on the lesion contrast of PET-positive liver metastases in cDWI and vcDWI. Methods We performed a retrospective analysis of 10 patients (4 m, 63.5 ± 12.9 y/o) with PET-positive liver metastases examined in 3 T-PET/MRI with b = 100,600,800,1000 and 1500s/mm2. cDWI (cb1000/cb1500) and vcDWI were computed based on following combinations: i) b = 100/600 s/mm2, ii) b = 100/800 s/mm2, iii) b = 100/1000s/mm2, iv) b = 100/600/1000s/mm2 v) all measured b-values. Mean signal intensity (SI) and standard deviation (SD) in the liver, spleen, kidney, bone marrow and in liver lesions were acquired. The coefficient of variation (CV = SD/SI), the differences of SI between measured and calculated high b-value images and the lesion contrast (SI lesion/liver) were computed. Results With increasing upper measured b-values, the CV in cDWI and vcDWI decreased (CV in the liver in cb1500: 0.42 with b100/600 s/mm2 and 0.28 with b100/b1000s/mm2) while the differences of measured and calculated b-value images decreased (in the liver in cb1500: 30.7% with b = 100/600 s/mm2, 19.7% with b100/b1000s/mm2). In diffusion-restricted lesions, lesion contrast was at least 1.6 in cb1000 and 1.4 in cb1500, respectively, with an upper measured b-value of b = 800 s/mm2 and 2.1 for vcDWI with an upper measured b-value of b = 1000s/mm2. Overall, the lesion contrast was superior in cb1500 and vcDWI compared to cb1000 (15% and 11%, respectively). Conclusion Measuring higher upper b-values seems to lead to more precise computed high b-value images and a decrease of CV. vcDWI provides a comparable lesion contrast to b = 1500s/mm2 and offers additionally the reduction of T2 shine-through effects. For vcDWI, measuring b = 1000s/mm2 as upper b-value seems to be necessary to guarantee good lesion visibility in the liver based on our preliminary results.
Collapse
|
40
|
Role of Non-Functional Imaging in the Diagnosis of Abdominal Neuroendocrine Tumors. Updates Surg 2018. [DOI: 10.1007/978-88-470-3955-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Manoharan J, Albers MB, Bartsch DK. The future: diagnostic and imaging advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer 2017; 24:T209-T225. [PMID: 28790162 DOI: 10.1530/erc-17-0231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Prospective randomized data are lacking, but current clinical expert guidelines recommend annual screening examinations, including laboratory assessments and various imaging modalities (e.g. CT, MRI, scintigraphy and EUS) for patients with multiple endocrine neoplasia type 1 (MEN1). Routine screening is proposed to detect and localize neuroendocrine manifestations as early as possible. The goal is timely intervention to improve quality of life and to increase life expectancy by preventing the development of life-threatening hormonal syndromes and/or metastatic disease. In recent years, some studies compared different and new imaging methods regarding their sensitivity and utility in MEN1 patients. This present article reviews the proposed diagnostic tools for MEN1 screening as well as potential future perspectives.
Collapse
Affiliation(s)
- Jerena Manoharan
- Department of VisceralThoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Max B Albers
- Department of VisceralThoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of VisceralThoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
42
|
Michael M, Garcia-Carbonero R, Weber MM, Lombard-Bohas C, Toumpanakis C, Hicks RJ. The Antiproliferative Role of Lanreotide in Controlling Growth of Neuroendocrine Tumors: A Systematic Review. Oncologist 2017; 22:272-285. [PMID: 28220021 PMCID: PMC5344642 DOI: 10.1634/theoncologist.2016-0305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neuroendocrine tumors (NETs) are a heterogeneous group of tumors, with >50% of cases involving the gastrointestinal system or pancreas. Somatostatin analogs (SSAs) are used for treating NET-related secretory syndromes and, more recently, for their antiproliferative effects. We conducted a systematic review of published literature on the antiproliferative efficacy and safety of the SSA lanreotide Autogel in the management of NETs to gain a fuller understanding of the evidence and identify future areas of research. METHODS Searches were conducted in PubMed up to March 16, 2016, and in the proceedings of four congresses from 2013 to 2016. RESULTS Screening of 1,132 publications identified in the searches found 40 relevant publications, including 27 full-length publications and 13 congress abstracts. Twenty-four of these publications reported antiproliferative efficacy data for lanreotide Autogel. The CLARINET study showed that 120 mg lanreotide Autogel every 4 weeks improves progression-free survival (PFS) in patients with gastroenteropancreatic (GEP)-NETs, with grade 1 or grade 2 (Ki-67 <10%) disease, providing class I evidence of its antiproliferative effects. The CLARINET open-label extension study reported a median PFS of 32.8 months with lanreotide Autogel. Other smaller studies generally support CLARINET. CONCLUSION Current clinical evidence shows that lanreotide Autogel has good antiproliferative activity with favorable safety and tolerability in patients with GEP-NETs, suggesting it should be considered as an early first-line treatment in this population. Further studies are needed to assess the potential benefits of higher doses and the use of lanreotide Autogel in combination therapy and as maintenance therapy in the absence of disease progression following other therapies. The Oncologist 2017;22:272-285 IMPLICATIONS FOR PRACTICE: This review presents the current clinical evidence for the antiproliferative activity of lanreotide Autogel in patients with midgut or pancreatic neuroendocrine tumors (NETs) and shows its effectiveness, safety, and tolerability in these patient populations. By systematically presenting all the clinical evidence, the review adds to existing publications by discussing results in a broad range of settings. The review also indicates future directions for investigation of the use of lanreotide Autogel in NETs originating in other locations, in combination therapy, or as maintenance therapy in progressive disease.
Collapse
Affiliation(s)
- Michael Michael
- Neuorendocrine Service & Division of Cancer Medicine, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Australia
| | | | | | | | | | - Rodney J Hicks
- Cancer Imaging & Neuroendocrine Service & Molecular Imaging and Targeted Therapeutics Laboratory, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, Australia
| |
Collapse
|
43
|
Hendifar AE, Marchevsky AM, Tuli R. Neuroendocrine Tumors of the Lung: Current Challenges and Advances in the Diagnosis and Management of Well-Differentiated Disease. J Thorac Oncol 2016; 12:425-436. [PMID: 27890494 DOI: 10.1016/j.jtho.2016.11.2222] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/31/2022]
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of malignancies that arise from neuroendocrine cells throughout the body, most commonly originating from the lungs and gastrointestinal tract. Lung NETs can be classified as well differentiated (low-grade typical carcinoids [TCs] and intermediate-grade atypical carcinoids [ACs]) and poorly differentiated (high-grade large cell neuroendocrine carcinoma or SCLC). The incidence of these tumors is increasing, but disease awareness remains low among thoracic specialists, who are often involved in the diagnosis and early treatment for these patients. An accurate and timely diagnosis can ensure the implementation of appropriate treatment and have a substantial impact on prognosis. However, lung NET classification and diagnosis, particularly for TCs/ACs, are complicated by several factors, including a variable natural history and nonspecific symptoms. Surgery remains the only curative option for TCs/ACs, but there is a lack of consensus between lung NET management guidelines regarding optimal treatment approaches in the unresectable/metastatic setting on account of the limited availability of high-level clinical evidence. As a result, a multidisciplinary approach to management of lung NETs is required to ensure a consistent and optimal level of care. RADIANT-4 is the first phase III trial involving a large subpopulation of patients with advanced well-differentiated lung NETs to report reductions in the risk for disease progression and death with everolimus over placebo. This led to the recent U.S. approval of everolimus-the first agent approved for advanced lung TCs/ACs. To further improve evidence-based care, additional randomized controlled trials in patients with lung carcinoids are needed.
Collapse
Affiliation(s)
- Andrew E Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Alberto M Marchevsky
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Richard Tuli
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
44
|
Cazzato RL, Garnon J, Ramamurthy N, Tsoumakidou G, Imperiale A, Namer IJ, Bachellier P, Caudrelier J, Rao P, Koch G, Gangi A. 18F-FDOPA PET/CT-Guided Radiofrequency Ablation of Liver Metastases from Neuroendocrine Tumours: Technical Note on a Preliminary Experience. Cardiovasc Intervent Radiol 2016; 39:1315-21. [PMID: 27048487 DOI: 10.1007/s00270-016-1334-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/26/2016] [Indexed: 12/20/2022]
Abstract
AIM To review our preliminary experience with 6-L-18F-fluorodihydroxyphenylalanine (18F-FDOPA) PET/CT-guided radiofrequency ablation (RFA) of liver metastases from neuroendocrine tumours (NETs). MATERIALS AND METHODS Three patients (mean age 51.3 years; range 43-56) with gastro-entero pancreatic NET (GEP-NET) liver metastases underwent 18F-FDOPA PET/CT-guided RFA. Patients were referred with oligometastatic hepatic-confined disease (1-6 metastases; <3 cm) on 18F-FDOPA PET/CT; poor lesion visualisation on US, CT, and MR; and ongoing symptoms. Procedures were performed in an interventional PET/CT scanner under general anaesthesia using a split-dose protocol. Lesion characteristics, procedural duration and technical success (accurate probe placement and post-procedural ablation-zone photopaenia), complications, patient and operator dose, and clinical outcomes were evaluated. RESULTS Thirteen liver metastases (mean size 11.4 mm, range 8-16) were treated in three patients (two presented with "carcinoid syndrome"). Technical success was 100 % with a mean procedural duration of 173.3 min (range 90-210) and no immediate complications. Mean patient dose was 2844 mGy·cm (range 2104-3686). Operator and radiographer doses were acceptable other than the operator's right hand in the first case (149 µSv); this normalised in the second case. There was no local tumour or extra-hepatic disease progression at mid-term follow-up (mean 12.6 months; range 6-20); however, two cases progressed with new liver metastases at different sites. There was 100 % clinical success (n = 2) in resolving carcinoid syndrome symptoms. CONCLUSION 18F-FDOPA PET/CT-guided RFA appears technically feasible, safe, and effective in patients with GEP-NETs and low-burden hepatic metastases. Further prospective studies are required to elucidate its precise role in tailored multimodality management of GEP-NET liver metastases.
Collapse
Affiliation(s)
- Roberto Luigi Cazzato
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France.
| | - Julien Garnon
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| | - Nitin Ramamurthy
- Department of Radiology, Norfolk and Norwich University Hospital, Colney Lane, Norwich, NR4 7UY, UK
| | - Georgia Tsoumakidou
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| | - Alessio Imperiale
- Department of Biophysics and Nuclear Medicine, Hôpital de Hautepierre (Hôpitaux Universitaires de Strasbourg, HUS), Avenue Molière, 67200, Strasbourg, France
| | - Izzie Jacques Namer
- Department of Biophysics and Nuclear Medicine, Hôpital de Hautepierre (Hôpitaux Universitaires de Strasbourg, HUS), Avenue Molière, 67200, Strasbourg, France
| | - Philippe Bachellier
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Hôpital de Hautepierre (Hôpitaux Universitaires de Strasbourg, HUS), Avenue Molière, 67200, Strasbourg, France
| | - Jean Caudrelier
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| | - Pramod Rao
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| | - Guillaume Koch
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| | - Afshin Gangi
- Department of Interventional Radiology, Nouvel Hôpital Civil (Hôpitaux Universitaires de Strasbourg, HUS), 1, place de l'Hôpital, 67000, Strasbourg, France
| |
Collapse
|