1
|
Di Franco M, Lamberti G, Campana D, Ambrosini V. Molecular Imaging for Response Assessment of Neuroendocrine Tumors (NET). Semin Nucl Med 2025:S0001-2998(25)00049-2. [PMID: 40345899 DOI: 10.1053/j.semnuclmed.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025]
Abstract
Assessing treatment response in neuroendocrine tumors (NET) remains a significant challenge due to their typically indolent growth and heterogenity, the frequent occurrence of disease stabilization rather than tumor shrinkage after therapy, and the inherent limitations of conventional imaging criteria. While molecular imaging-primarily somatostatin receptor (SST) PET/CT-has improved lesion detection, the absence of standardized response criteria limits its clinical utility and prevents its use as full replacement of conventional imaging. Emerging strategies, including revised thresholds for dimensional changes, criteria evaluating different features, such as lesions' density and functional tumor volumes, offer potential improvements in response evaluation but require further validation for routine clinical implementation. This review examines the current challenges in assessing NET treatment response, evaluates the strengths and limitations of available imaging modalities, and discusses emerging approaches and future directions for optimizing therapeutic monitoring in the heterogeneous panorama of NET.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy.
| | - Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Davide Campana
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy; Medical Oncology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy; Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Megherbi I, Hoog C, Perrier M, Brixi H, Cadiot G, Hoeffel-Fornes C, Morland D. Quantitative 177Lu-DOTATATE SPECT/CT is predictive of 6-month PRRT morphological response in midgut neuroendocrine tumors: a pilot study. EJNMMI Res 2025; 15:53. [PMID: 40329099 PMCID: PMC12055673 DOI: 10.1186/s13550-025-01250-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND peptide receptor radionuclide therapy with 177Lu-DOTATATE has become an established second-line treatment for patients with advanced small intestine neuroendocrine tumors (siNET). Treatment efficacy is assessed several months after the end of treatment and is based on RECIST criteria. Post-therapy scintigraphy (PTS) can be performed after each cycle, but its value in early response assessment is debated particularly given the lack of quantification available in clinical routine. New quantification modules are now available, enabling automatic SUV calculation. The main goal of this study is to assess the value of the evolution of SUV between the first (C1) and the second (C2) cycle on quantitative PTS in predicting response to treatment. All patients with siNET referred to our center for treatment with 177Lu-DOTATATE were included. The SUVmax of the lesion with the greatest uptake was measured on PTS SPECT/CT at C1 and C2. ∆SUVmax was calculated. Linear regression between ∆SUVmax and 6-month RECIST percentage was used. Relative changes in tumor metabolic volume (MTV) were also studied along with clinical parameters. RESULTS twelve consecutive patients with progressive metastatic siNET were included. One patient showed partial response at 6 months, the other were considered as stable disease. Linear regression showed that 6-month RECIST percentage and ∆SUVmax were strongly correlated with the following regression formula: 6-month RECIST = -0.05 + 0.36 x ∆SUVmax (p < 0.001). ∆MTV was not predictive of response. CONCLUSION we report a significant link between PTS ∆SUVmax and 6-month RECIST percentage in patients with siNET. Quantitative imaging would thus enable the prediction of 6-month response of 177Lu-DOTATATE as early as C2. These results need to be confirmed on a larger population.
Collapse
Affiliation(s)
| | | | - Marine Perrier
- Gastroentérologie et Oncologie digestive, CHU de Reims, Reims, France
| | - Hedia Brixi
- Gastroentérologie et Oncologie digestive, CHU de Reims, Reims, France
| | - Guillaume Cadiot
- Gastroentérologie et Oncologie digestive, CHU de Reims, Reims, France
| | - Christine Hoeffel-Fornes
- Radiologie, CHU de Reims, Reims, France
- CReSTIC, Université de Reims Champagne-Ardenne, Reims, 3804, UR, France
| | - David Morland
- Médecine Nucléaire, Institut Godinot, Reims, France.
- CReSTIC, Université de Reims Champagne-Ardenne, Reims, 3804, UR, France.
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
3
|
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
|
4
|
Halperin R, Tirosh A. Mid-Treatment Response to 177-Lutetium Dotatate Predicts Overall Outcome in Patients With Advanced Neuroendocrine Tumors. JCO Oncol Pract 2024; 20:1401-1409. [PMID: 38935916 DOI: 10.1200/op.23.00789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 06/29/2024] Open
Abstract
PURPOSE Patients with advanced, well-differentiated neuroendocrine tumors (WD-NETs) often require both peptide receptor radionuclide therapy (PRRT) and subsequent chemotherapy. However, no mid-PRRT predictors are available to identify patients who will not benefit from subsequent PRRT to limit their radiation exposure. Our aim is to characterize patients for whom subsequent PRRT is less efficacious on the basis of mid-PRRT evaluation. MATERIALS AND METHODS A retrospective study of patients with WD-NET who underwent ≥four PRRT cycles. Data gathered included demographics, tumor grade, stage, and response (partial response [PR], stable disease [SD], and progressive disease [PD]) on the basis of RECIST 1.1 criteria and 68Ga-dotatate positron emission tomography-computerized tomography pretreatment, after second and fourth treatment cycle, 6 months after fourth cycle, and at last follow-up. RESULTS Fifty-one patients (51.6% women; age at diagnosis 66.0 ± 1.65 years), with pancreatic NET (PNET; n = 24), small intestine NET (n = 13), or other NET (n = 14), received PRRT, resulting in PR (n = 21), SD (n = 23), and PD (n = 3). Of the patients reaching PR after PRRT, most reached PR after two treatments (70.4%), with only 11.8% PR occurring between subsequent cycles (P = .001). Furthermore, patients with PR at mid-treatment had higher PR rates after PRRT completion than those with SD (P = .007). Patients harboring PNET who achieved PR had a more pronounced reduction of tumor burden in additional cycles than patients who did not (25.6% v 1.5%; P = .03, respectively). On the multivariable model, adjusted for grade and primary site, PR at mid-treatment evaluation was associated with a 20.9 adjusted odds ratio for additional PR at PRRT completion (P = .002). CONCLUSION Mid-PRRT assessment predicts subsequent PRRT response in patients with WD-NET, especially those with PNET, informing personalized management and consideration of reduced bone marrow radiation exposure in high-risk patients.
Collapse
Affiliation(s)
- Reut Halperin
- ENTIRE-Endocrine Neoplasia Translational Research Center, Ramat Gan, Israel
- Division of Endocrinology, Metabolism and Diabetes, Sheba Medical Center, Ramat Gan, Israel
- Tel Aviv University Faculty of Medicine, Ramat Gan, Israel
| | - Amit Tirosh
- ENTIRE-Endocrine Neoplasia Translational Research Center, Ramat Gan, Israel
- Division of Endocrinology, Metabolism and Diabetes, Sheba Medical Center, Ramat Gan, Israel
- Tel Aviv University Faculty of Medicine, Ramat Gan, Israel
| |
Collapse
|
5
|
Scalorbi F, Garanzini EM, Calareso G, Marzi C, Di Rocco G, Argiroffi G, Baccini M, Pusceddu S, Marchianò A, Maccauro M. Cylindrical TGR as early radiological predictor of RLT progression in GEPNETs: a proof of concept. Sci Rep 2024; 14:15782. [PMID: 38982134 PMCID: PMC11233714 DOI: 10.1038/s41598-024-66668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
This study aims to assess the predictive capability of cylindrical Tumor Growth Rate (cTGR) in the prediction of early progression of well-differentiated gastro-entero-pancreatic tumours after Radio Ligand Therapy (RLT), compared to the conventional TGR. Fifty-eight patients were included and three CT scans per patient were collected at baseline, during RLT, and follow-up. RLT response, evaluated at follow-up according to RECIST 1.1, was calculated as a percentage variation of lesion diameters over time (continuous values) and as four different RECIST classes. TGR between baseline and interim CT was computed using both conventional (approximating lesion volume to a sphere) and cylindrical (called cTGR, approximating lesion volume to an elliptical cylinder) formulations. Receiver Operating Characteristic (ROC) curves were employed for Progressive Disease class prediction, revealing that cTGR outperformed conventional TGR (area under the ROC equal to 1.00 and 0.92, respectively). Multivariate analysis confirmed the superiority of cTGR in predicting continuous RLT response, with a higher coefficient for cTGR (1.56) compared to the conventional one (1.45). This study serves as a proof of concept, paving the way for future clinical trials to incorporate cTGR as a valuable tool for assessing RLT response.
Collapse
Affiliation(s)
- Federica Scalorbi
- Nuclear Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Enrico Matteo Garanzini
- Department of Radiodiagnostics and Radiotherapy, IRCCS Fondazione Istituto Nazionale Tumori, Milan, Italy
| | - Giuseppina Calareso
- Department of Radiodiagnostics and Radiotherapy, IRCCS Fondazione Istituto Nazionale Tumori, Milan, Italy
| | - Chiara Marzi
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Viale Morgagni 59, 50134, Florence, Italy.
| | - Gabriella Di Rocco
- Department of Radiodiagnostics and Radiotherapy, IRCCS Fondazione Istituto Nazionale Tumori, Milan, Italy
- Post-Graduation School of Radiology, Department of Health Sciences, University of Milan, Milan, Italy
| | - Giovanni Argiroffi
- Nuclear Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Michela Baccini
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Viale Morgagni 59, 50134, Florence, Italy
| | - Sara Pusceddu
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfonso Marchianò
- Department of Radiodiagnostics and Radiotherapy, IRCCS Fondazione Istituto Nazionale Tumori, Milan, Italy
| | - Marco Maccauro
- Nuclear Medicine Department, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| |
Collapse
|
6
|
Padmanabhan Nair Sobha R, Jensen CT, Waters R, Calimano-Ramirez LF, Virarkar MK. Appendiceal Neuroendocrine Neoplasms: A Comprehensive Review. J Comput Assist Tomogr 2024; 48:545-562. [PMID: 37574653 DOI: 10.1097/rct.0000000000001528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
ABSTRACT Appendiceal neuroendocrine neoplasm (NEN) is the most common adult appendiceal malignant tumor, constituting 16% of gastrointestinal NENs. They are versatile tumors with varying morphology, immunohistochemistry, secretory properties, and cancer genomics. They are slow growing and clinically silent, to begin with, or present with features of nonspecific vague abdominal pain. Most acute presentations are attributed clinically to appendicitis, with most cases detected incidentally on pathology after an appendectomy. Approximately 40% of them present clinically with features of hormonal excess, which is likened to the functional secretory nature of their parent cell of origin. The symptoms of carcinoid syndrome render their presence clinically evident. However, slow growing and symptomatically silent in its initial stages, high-grade neuroendocrine tumors and neuroendocrine carcinomas of the appendix are aggressive and usually have hepatic and lymph node metastasis at presentation. This review article focuses on imaging characteristics, World Health Organization histopathological classification and grading, American Joint Committee on Cancer/Union or International Cancer Control, European Neuroendocrine Tumor Society staging, European Neuroendocrine Tumor Society standardized guidelines for reporting, data interpretation, early-stage management protocols, and advanced-stage appendiceal NENs. Guidelines are also set for the follow-up and reassessment. The role of targeted radiotherapy, chemotherapy, and high-dose somatostatin analogs in treating advanced disease are discussed, along with types of ablative therapies and liver transplantation for tumor recurrence. The search for newer location-specific biomarkers in NEN is also summarized. Regarding the varying aggressiveness of the tumor, there is a scope for research in the field, with plenty of data yet to be discovered.
Collapse
Affiliation(s)
| | - Corey T Jensen
- From the Department of Radiology, University of Texas MD Anderson Cancer Center
| | - Rebecca Waters
- Department of Pathology and Lab Medicine MD Anderson Cancer Center, Houston, TX
| | | | - Mayur K Virarkar
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| |
Collapse
|
7
|
Di Franco M, Zanoni L, Fortunati E, Fanti S, Ambrosini V. Radionuclide Theranostics in Neuroendocrine Neoplasms: An Update. Curr Oncol Rep 2024; 26:538-550. [PMID: 38581469 PMCID: PMC11063107 DOI: 10.1007/s11912-024-01526-5] [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] [Accepted: 03/26/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE OF REVIEW This paper aims to address the latest findings in neuroendocrine tumor (NET) theranostics, focusing on new evidence and future directions of combined diagnosis with positron emission tomography (PET) and treatment with peptide receptor radionuclide therapy (PRRT). RECENT FINDINGS Following NETTER-1 trial, PRRT with [177Lu]Lu-DOTATATE was approved by FDA and EMA and is routinely employed in advanced G1 and G2 SST (somatostatin receptor)-expressing NET. Different approaches have been proposed so far to improve the PRRT therapeutic index, encompassing re-treatment protocols, combinations with other therapies and novel indications. Molecular imaging holds a potential added value in characterizing disease biology and heterogeneity using different radiopharmaceuticals (e.g., SST and FDG) and may provide predictive and prognostic parameters. Response assessment criteria are still an unmet need and new theranostic pairs showed preliminary encouraging results. PRRT for NET has become a paradigm of modern theranostics. PRRT holds a favorable toxicity profile, and it is associated with a prolonged time to progression, reduction of symptoms, and improved patients' quality of life. In light of further optimization, different new strategies have been investigated, along with the development of new radiopharmaceuticals.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Lucia Zanoni
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Emilia Fortunati
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
8
|
Ha S, Kim YI, Oh JS, Yoo C, Ryoo BY, Ryu JS. Prediction of [ 177Lu]Lu-DOTA-TATE therapy response using the absorbed dose estimated from [ 177Lu]Lu-DOTA-TATE SPECT/CT in patients with metastatic neuroendocrine tumour. EJNMMI Phys 2024; 11:14. [PMID: 38315270 PMCID: PMC10844176 DOI: 10.1186/s40658-024-00620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Peptide receptor radionuclide therapy (PRRT) with [177Lu]Lu-DOTA-TATE has shown efficacy in patients with metastatic neuroendocrine tumours (NETs). Personalised dosimetry is crucial to optimise treatment outcomes and minimise adverse events. In this study, we investigated the correlation between the tumour-absorbed dose (TAD) estimated from [177Lu]Lu-DOTA-TATE SPECT/CT and the therapeutic response. METHOD A retrospective analysis was conducted on patients with advanced well-differentiated NETs grades 1-3 who underwent PRRT and exhibited greater uptake than liver on pre-therapeutic [68Ga]Ga-DOTA-TOC PET/CT. Target lesions were selected based on the RECIST 1.1 and PERCIST 1.0 criteria using [177Lu]Lu-DOTA-TATE SPECT/CT and pre-therapeutic contrast-enhanced CT scans. For anatomical image analysis, the sum of the longest diameter (SLD) of the target lesions was measured using the RECIST 1.1 criteria for patient-based analysis and the longest diameter (LD) of the target lesion using the RECIST-L criteria for lesion-based analysis. Standardised uptake values (SUVs) were measured on SPECT/CT images, and TADs were calculated based on the SUVs. Dosimetry was performed using a single SPECT/CT imaging time point at day 4-5 post-therapy. Statistical analyses were conducted to investigate correlations and determine the target lesion responses. RESULTS Twenty patients with primary tumour sites and hepatic metastases were included. Fifty-five target lesions, predominantly located in the pancreas and liver, were analysed. The cumulative TAD (lesion-based analysis: r = 0.299-0.301, p = 0.025-0.027), but not the cycle 1 SUV (lesion-based analysis: r = 0.198-0.206, p = 0.131-0.147) or cycle 1 TAD (lesion-based analysis: r = 0.209-0.217, p = 0.112-0.126), exhibited a significant correlation with the change in LD of the target lesion. Binary logistic regression analysis identified the significance of the cumulative TAD in predicting disease control according to the RECIST-L criteria (odds ratio = 1.031-1.051, p = 0.024-0.026). CONCLUSIONS The cumulative TAD estimated from [177Lu]Lu-DOTA-TATE SPECT/CT revealed a significant correlation with change in LD, which was significantly higher for the cumulative TAD than for the cycle 1 SUV or TAD. A higher cumulative TAD was associated with disease control in the target lesion. However, considering the limitations inherent to a confined sample size, careful interpretation of these findings is required. Estimation of the cumulative TAD of [177Lu]Lu-DOTA-TATE therapy could guide the platform towards personalised therapy.
Collapse
Affiliation(s)
- Sejin Ha
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong-Il Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea.
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
| | - Changhoon Yoo
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Baek-Yeol Ryoo
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Sook Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Theranostics Center, Asan Cancer Institute, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
9
|
Yazdani E, Geramifar P, Karamzade-Ziarati N, Sadeghi M, Amini P, Rahmim A. Radiomics and Artificial Intelligence in Radiotheranostics: A Review of Applications for Radioligands Targeting Somatostatin Receptors and Prostate-Specific Membrane Antigens. Diagnostics (Basel) 2024; 14:181. [PMID: 38248059 PMCID: PMC10814892 DOI: 10.3390/diagnostics14020181] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Radiotheranostics refers to the pairing of radioactive imaging biomarkers with radioactive therapeutic compounds that deliver ionizing radiation. Given the introduction of very promising radiopharmaceuticals, the radiotheranostics approach is creating a novel paradigm in personalized, targeted radionuclide therapies (TRTs), also known as radiopharmaceuticals (RPTs). Radiotherapeutic pairs targeting somatostatin receptors (SSTR) and prostate-specific membrane antigens (PSMA) are increasingly being used to diagnose and treat patients with metastatic neuroendocrine tumors (NETs) and prostate cancer. In parallel, radiomics and artificial intelligence (AI), as important areas in quantitative image analysis, are paving the way for significantly enhanced workflows in diagnostic and theranostic fields, from data and image processing to clinical decision support, improving patient selection, personalized treatment strategies, response prediction, and prognostication. Furthermore, AI has the potential for tremendous effectiveness in patient dosimetry which copes with complex and time-consuming tasks in the RPT workflow. The present work provides a comprehensive overview of radiomics and AI application in radiotheranostics, focusing on pairs of SSTR- or PSMA-targeting radioligands, describing the fundamental concepts and specific imaging/treatment features. Our review includes ligands radiolabeled by 68Ga, 18F, 177Lu, 64Cu, 90Y, and 225Ac. Specifically, contributions via radiomics and AI towards improved image acquisition, reconstruction, treatment response, segmentation, restaging, lesion classification, dose prediction, and estimation as well as ongoing developments and future directions are discussed.
Collapse
Affiliation(s)
- Elmira Yazdani
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran 14117-13135, Iran
| | - Najme Karamzade-Ziarati
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran 14117-13135, Iran
| | - Mahdi Sadeghi
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Payam Amini
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
| |
Collapse
|
10
|
Siripongsatian D, de Lussanet de la Sablonière QG, Anton Verburg F, Brabander T. How to design a theranostic trial? ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2024; 4:e230045. [PMID: 38770190 PMCID: PMC11103757 DOI: 10.1530/eo-23-0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/17/2024] [Indexed: 05/22/2024]
Abstract
The field of nuclear theranostic clinical trials is continuously expanding as an increasing number of novel agents and treatment combinations are explored for treating advanced and metastatic cancers. Moving from 'bench-to-bedside' is oftentimes a complex and lengthy process. The objective of this overview is to explore the basic elements involved in designing clinical trials with a special focus on theranostics in nuclear medicine. The 'bench-to-bedside' journey involves translating basic scientific research into patient-effective treatments. Preclinical studies, a crucial initial step, are a complex process encompassing in vitro experiments, in vivo studies, and animal models to explore hypotheses in humans. Clinical trials follow, with predefined phases assessing safety, effectiveness, and comparisons to existing treatments. This process demands investments in data management, statistics, good clinical practice (GCP) accreditations, and collaborative efforts for funding and sustainable pricing. Theranostics, merging diagnostics and personalized treatment, is at the forefront. Continuous efforts to enhance existing agents involve reducing adverse effects, exploring new indications, and incorporating advanced imaging modalities. Radionuclide therapy, unique with non-uniform distribution and complex radiobiology, plays a distinct role. This article explores trends and challenges in each clinical trial phase in light of the emerging field of theranostics in nuclear medicine, emphasizing meticulous trial design, dosimetry optimization, and the necessity of collaborative stakeholder efforts for successful implementation.
Collapse
Affiliation(s)
| | | | | | - Tessa Brabander
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
11
|
Lee H, Kipnis ST, Niman R, O’Brien SR, Eads JR, Katona BW, Pryma DA. Prediction of 177Lu-DOTATATE Therapy Outcomes in Neuroendocrine Tumor Patients Using Semi-Automatic Tumor Delineation on 68Ga-DOTATATE PET/CT. Cancers (Basel) 2023; 16:200. [PMID: 38201627 PMCID: PMC10778298 DOI: 10.3390/cancers16010200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Treatment of metastatic neuroendocrine tumors (NET) with 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) results in favorable response only in a subset of patients. We investigated the prognostic value of quantitative pre-treatment semi-automatic 68Ga-DOTATATE PET/CT analysis in NET patients treated with PRRT. METHODS The medical records of 94 NET patients who received at least one cycle of PRRT at a single institution were retrospectively reviewed. On each pre-treatment 68Ga-DOTATATE PET/CT, the total tumor volume (TTV), maximum tumor standardized uptake value for the patient (SUVmax), and average uptake in the lesion with the lowest radiotracer uptake (SUVmin) were determined with a semi-automatic tumor delineation method. Progression-free survival (PFS) and overall survival (OS) among the patients were compared based on optimal cutoff values for the imaging parameters. RESULTS On Kaplan-Meier analysis and univariate Cox regression, significantly shorter PFS was observed in patients with lower SUVmax, lower SUVmin, and higher TTV. On multivariate Cox regression, lower SUVmin and higher TTV remained predictive of shorter PFS. Only higher TTV was found to be predictive of shorter OS on Kaplan-Meier and Cox regression analyses. In a post hoc Kaplan-Meier analysis, patients with at least one high-risk feature (low SUVmin or high TTV) showed shorter PFS and OS, which may be the most convenient parameter to measure in clinical practice. CONCLUSIONS The tumor volume and lowest lesion uptake on 68Ga-DOTATATE PET/CT can predict disease progression following PRRT in NET patients, with the former also predictive of overall survival. NET patients at risk for poor outcomes following PRRT can be identified with semi-automated quantitative analysis of 68Ga-DOTATATE PET/CT.
Collapse
Affiliation(s)
- Hwan Lee
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sarit T. Kipnis
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Medicine, Georgetown University, Washington, DC 20007, USA
| | - Remy Niman
- MIM Software Inc., Cleveland, OH 44122, USA
| | - Sophia R. O’Brien
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jennifer R. Eads
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bryson W. Katona
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel A. Pryma
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Wetz C, Ruhwedel T, Schatka I, Grabowski J, Jann H, Metzger G, Galler M, Amthauer H, Rogasch JMM. Plasma Markers for Therapy Response Monitoring in Patients with Neuroendocrine Tumors Undergoing Peptide Receptor Radionuclide Therapy. Cancers (Basel) 2023; 15:5717. [PMID: 38136263 PMCID: PMC10741556 DOI: 10.3390/cancers15245717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Pretherapeutic chromogranin A, alkaline phosphatase (ALP), or De Ritis ratio (aspartate aminotransferase/alanine aminotransferase) are prognostic factors in patients with metastatic neuroendocrine tumors (NET) undergoing peptide receptor radionuclide therapy (PRRT). However, their value for intratherapeutic monitoring remains unclear. We evaluated if changes in plasma markers during PRRT can help identify patients with unfavorable outcomes. METHODS A monocentric retrospective analysis of 141 patients with NET undergoing PRRT with [177Lu]Lu-DOTATOC was conducted. Changes in laboratory parameters were calculated by dividing the values determined immediately before each cycle of PRRT by the pretherapeutic value. Patients with low vs. high PFS were compared with the Wilcoxon rank-sum test. RESULTS Progression, relapse, or death after PRRT was observed in 103/141 patients. Patients with low PFS showed a significant relative ALP increase before the third (p = 0.014) and fourth (p = 0.039) cycles of PRRT. Kaplan-Meier analysis revealed a median PFS of 24.3 months (95% CI, 20.7-27.8 months) in patients with decreasing ALP values (Δ > 10%) during treatment, 12.5 months (95% CI, 9.2-15.8 months) in patients with increasing ALP values (Δ > 10%), and 17.7 months (95% CI, 13.6-21.8 months) with stable ALP values (Δ ± 10%). CONCLUSIONS Based on these exploratory data, a rise in plasma ALP might indicate disease progression and should be interpreted cautiously during therapy.
Collapse
Affiliation(s)
- Christoph Wetz
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Tristan Ruhwedel
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Imke Schatka
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Jane Grabowski
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Henning Jann
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Giulia Metzger
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Markus Galler
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
| | - Julian M. M. Rogasch
- Department of Nuclear Medicine, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.R.); (G.M.); (M.G.); (H.A.); (J.M.M.R.)
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
| |
Collapse
|
13
|
Mariën L, Islam O, Chhajlani S, Lybaert W, Peeters M, Van Camp G, Op de Beeck K, Vandamme T. The Quest for Circulating Biomarkers in Neuroendocrine Neoplasms: a Clinical Perspective. Curr Treat Options Oncol 2023; 24:1833-1851. [PMID: 37989978 DOI: 10.1007/s11864-023-01147-3] [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] [Accepted: 10/24/2023] [Indexed: 11/23/2023]
Abstract
OPINION STATEMENT Given the considerable heterogeneity in neuroendocrine neoplasms (NENs), it appears unlikely that a sole biomarker exists capable of fully capturing all useful clinical aspects of these tumors. This is reflected in the abundant number of biomarkers presently available for the diagnosis, prognosis, and monitoring of NEN patients. Although assessment of immunohistochemical and radiological markers remains paramount and often obligatory, there has been a notable surge of interest in circulating biomarkers over the years given the numerous benefits associated with liquid biopsies. Currently, the clinic primarily relies on single-analyte assays such as the chromogranin A assay, but these are far from ideal because of limitations such as compromised sensitivity and specificity as well as a lack of standardization. Consequently, the quest for NEN biomarkers continued with the exploration of multianalyte markers, exemplified by the development of the NETest and ctDNA-based analysis. Here, an extensive panel of markers is simultaneously evaluated to identify distinct signatures that could enhance the accuracy of patient diagnosis, prognosis determination, and response to therapy prediction and monitoring. Given the promising results, the development and implementation of these multianalyte markers are expected to usher in a new era of NEN biomarkers in the clinic. In this review, we will outline both clinically implemented and more experimental circulating markers to provide an update on developments in this rapidly evolving field.
Collapse
Affiliation(s)
- Laura Mariën
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
- Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Odeta Islam
- Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
- NETwerk and Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Siddharth Chhajlani
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
- NETwerk and Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Willem Lybaert
- NETwerk and Department of Oncology, VITAZ, Lodewijk de Meesterstraat 5, 9100, Sint-Niklaas, Belgium
| | - Marc Peeters
- Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
- NETwerk and Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650, Edegem, Belgium
- Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Timon Vandamme
- Integrated Personalized and Precision Oncology Network (IPPON), Center for Oncological Research (CORE), University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610, Wilrijk, Belgium.
- NETwerk and Department of Oncology, Antwerp University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium.
| |
Collapse
|
14
|
Halfdanarson TR, Mallak N, Paulson S, Chandrasekharan C, Natwa M, Kendi AT, Kennecke HF. Monitoring and Surveillance of Patients with Gastroenteropancreatic Neuroendocrine Tumors Undergoing Radioligand Therapy. Cancers (Basel) 2023; 15:4836. [PMID: 37835530 PMCID: PMC10571645 DOI: 10.3390/cancers15194836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Radioligand therapy (RLT) with [177Lu]Lu-DOTA-TATE is a standard of care for adult patients with somatostatin-receptor (SSTR)-positive gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Taking advantage of this precision nuclear medicine approach requires diligent monitoring and surveillance, from the use of diagnostic SSTR-targeted radioligand imaging for the selection of patients through treatment and assessments of response. Published evidence-based guidelines assist the multidisciplinary healthcare team by providing acceptable approaches to care; however, the sheer heterogeneity of GEP-NETs can make these frameworks difficult to apply in individual clinical circumstances. There are also contradictions in the literature regarding the utility of novel approaches in monitoring and surveilling patients with GEP-NETs receiving RLT. This article discusses the emerging evidence on imaging, clinical biochemistry, and tumor assessment criteria in the management of patients receiving RLT for GEP-NETs; additionally, it documents our own best practices. This allows us to offer practical guidance on how to effectively implement monitoring and surveillance measures to aid patient-tailored clinical decision-making.
Collapse
Affiliation(s)
| | - Nadine Mallak
- Division of Molecular Imaging and Therapy, Oregon Health and Science University, Portland, OR 97239, USA;
| | | | | | - Mona Natwa
- Langone Health, New York University, New York, NY 10016, USA
| | | | | |
Collapse
|
15
|
Piscopo L, Zampella E, Pellegrino S, Volpe F, Nappi C, Gaudieri V, Fonti R, Vecchio SD, Cuocolo A, Klain M. Diagnosis, Management and Theragnostic Approach of Gastro-Entero-Pancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2023; 15:3483. [PMID: 37444593 DOI: 10.3390/cancers15133483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Gastro-entero-pancreatic neuroendocrine neoplasms (GEP-NENs) constitute an ideal target for radiolabeled somatostatin analogs. The theragnostic approach is able to combine diagnosis and therapy by the identification of a molecular target that can be diagnosed and treated with the same radiolabeled compound. During the last years, advances in functional imaging with the introduction of somatostatin analogs and peptide receptor radionuclide therapy, have improved the diagnosis and treatment of GEP-NENs. Moreover, PET/CT imaging with 18F-FDG represents a complementary tool for prognostic evaluation of patients with GEP-NENs. In the field of personalized medicine, the theragnostic approach has emerged as a promising tool in diagnosis and management of patients with GEP-NENs. The aim of this review is to summarize the current evidence on diagnosis and management of patients with GEP-NENs, focusing on the theragnostic approach.
Collapse
Affiliation(s)
- Leandra Piscopo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Sara Pellegrino
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples, Federico II, 80131 Naples, Italy
| |
Collapse
|
16
|
Neuroendocrine Tumor Therapy Response Assessment. PET Clin 2023; 18:267-286. [PMID: 36858748 DOI: 10.1016/j.cpet.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Peptide receptor radionuclide therapy has become an integral part of management of neuroendocrine neoplasms. Gallium-68- and lutetium-177-labeled somatostatin receptor analogues have replaced yttrium-90- and 111-indium-based tracers. Several newer targeted therapies are also being used in clinical and research settings. It is imperative to accurately evaluate the response to these agents. The characteristics of NENs and the response patterns of the targeted therapies make response assessment in this group challenging. This article provides an overview of the strengths and weaknesses of the various biomarkers available for response assessment.
Collapse
|
17
|
Glucose Metabolism Modification Induced by Radioligand Therapy with [ 177Lu]Lu/[ 90Y]Y-DOTATOC in Advanced Neuroendocrine Neoplasms: A Prospective Pilot Study within FENET-2016 Trial. Pharmaceutics 2022; 14:pharmaceutics14102009. [PMID: 36297443 PMCID: PMC9612170 DOI: 10.3390/pharmaceutics14102009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022] Open
Abstract
[18F]F-FDG (FDG) PET is emerging as a relevant diagnostic and prognostic tool in neuroendocrine neoplasms (NENs), as a simultaneous decrease in [68Ga]Ga-DOTA peptides and increase in FDG uptake (the “flip-flop” phenomenon) occurs during the natural history of these tumors. The aim of this study was to evaluate the variations on FDG PET in NEN patients treated with two different schemes of radioligand therapy (RLT) and to correlate them with clinical−pathologic variables. A prospective evaluation of 108 lesions in 56 patients (33 males and 23 females; median age, 64.5 years) affected by NENs of various primary origins (28 pancreatic, 13 gastrointestinal, 9 bronchial, 6 unknown primary (CUP-NENs) and 1 pheochromocytoma) and grades (median Ki-67 = 9%) was performed. The patients were treated with RLT within the phase II clinical trial FENET-2016 (CTID: NCT04790708). RLT was offered for 32 patients with the MONO scheme (five cycles of [177Lu]Lu-DOTATOC) and for 24 with the DUO scheme (three cycles of [177Lu]Lu-DOTATOC alternated with two cycles of [90Y]Y-DOTATOC). Variations in terms of the ΔSUVmax of a maximum of three target lesions per patient (58 for MONO and 50 for DUO RLT) were assessed between baseline and 3 months post-RLT FDG PET. In patients with negative baseline FDG PET, the three most relevant lesions on [68Ga]Ga-DOTA-peptide PET were assessed and matched on post-RLT FDG PET, to check for any possible changes in FDG avidity. Thirty-five patients (62.5%) had at least one pathological FDG uptake at the baseline scans, but the number was reduced to 29 (52%) after RLT. In the patients treated with DUO-scheme RLT, 20 out of 50 lesions were FDG positive before therapy, whereas only 14 were confirmed after RLT (p = 0.03). Moreover, none of the 30 FDG-negative lesions showed an increased FDG uptake after RLT. The lesions of patients with pancreatic and CUP-NENs treated with the DUO scheme demonstrated a significant reduction in ΔSUVmax in comparison to those treated with MONO RLT (p = 0.03 and p = 0.04, respectively). Moreover, we found a mild positive correlation between the grading and ΔSUVmax in patients treated with the MONO scheme (r = 0.39, p < 0.02), while no evidence was detected for patients treated with the DUO scheme. Our results suggest that RLT, mostly with the DUO scheme, could be effective in changing NEN lesions’ glycometabolism, in particular, in patients affected by pancreatic and CUP-NENs, regardless of their Ki-67 index. Probably, associating [90Y]Y-labelled peptides, which have high energy emission and a crossfire effect, and [177Lu]Lu ones, characterized by a longer half-life and a safer profile for organs at risk, might represent a valid option in FDG-positive NENs addressed to RLT. Further studies are needed to validate our preliminary findings. In our opinion, FDG PET/CT should represent a potent tool for fully assessing a patient’s disease characteristics, both before and after RLT.
Collapse
|
18
|
Lee ONY, Tan KV, Tripathi V, Yuan H, Chan WWL, Chiu KWH. The Role of 68 Ga-DOTA-SSA PET/CT in the Management and Prediction of Peptide Receptor Radionuclide Therapy Response for Patients With Neuroendocrine Tumors : A Systematic Review and Meta-analysis. Clin Nucl Med 2022; 47:781-793. [PMID: 35485851 DOI: 10.1097/rlu.0000000000004235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this study was to identify and evaluate the role of 68 Ga-DOTA-somatostatin analog (SSA) PET/CT in guiding treatment for patients with neuroendocrine tumors (NETs) based on published literature, with specific focus on the ability of PET/CT to impact clinical management and predict peptide receptor radionuclide therapy (PRRT) response. PATIENTS AND METHODS A systematic literature search of articles up to December 2021 was performed using PubMed and Scopus. Eligible studies included ≥10 patients with confirmed or suspected NETs who had undergone pretreatment staging 68 Ga-DOTA-SSA PET/CT. A meta-analysis using the random-effects model was conducted to determine the overall change in management after PET/CT, whereas PET/CT-derived parameters that correlated with PRRT outcome were summarized from studies that assessed its predictive capabilities. RESULTS A total of 39 studies were included in this systemic review, of which 2266 patients from 24 studies were included for meta-analysis. We showed that PET/CT resulted in a change in clinical management in 36% (95% confidence interval, 31%-41%; range, 3%-66%) of patients. Fifteen studies consisting of 618 patients examined the prognostic ability of 68 Ga-DOTA-SSA PET/CT for PRRT. Of those, 8 studies identified a higher pretreatment SUV to favor PRRT, and 4 identified PET-based radiomic features for somatostatin receptor heterogeneity to be predictive of PRRT response. CONCLUSIONS Along with its diagnostic abilities, 68 Ga-DOTA-SSA PET/CT can impact treatment decision-making and may predict PRRT response in patients with NETs. More robust studies should be conducted to better elucidate the prognostic role of somatostatin receptor PET/CT in optimizing treatment for clinical outcome.
Collapse
Affiliation(s)
- Osher Ngo Yung Lee
- From the Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kel Vin Tan
- Department of Oncology, The University of Oxford, Oxford, United Kingdom
| | - Vrijesh Tripathi
- Department of Mathematics and Statistics, The University of the West Indies, St. Augustine Campus, Trinidad and Tobago
| | - Hui Yuan
- Department of Nuclear Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | - Keith Wan Hang Chiu
- Department of Diagnostic and Interventional Radiology, Kwong Wah Hospital, Hong Kong
| |
Collapse
|
19
|
Molecular and Anatomic Imaging of Neuroendocrine Tumors. Surg Oncol Clin N Am 2022; 31:649-671. [DOI: 10.1016/j.soc.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
20
|
Iravani A, Parihar AS, Akhurst T, Hicks RJ. Molecular imaging phenotyping for selecting and monitoring radioligand therapy of neuroendocrine neoplasms. Cancer Imaging 2022; 22:25. [PMID: 35659779 PMCID: PMC9164531 DOI: 10.1186/s40644-022-00465-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/26/2022] [Indexed: 11/10/2022] Open
Abstract
Neuroendocrine neoplasia (NEN) is an umbrella term that includes a widely heterogeneous disease group including well-differentiated neuroendocrine tumours (NETs), and aggressive neuroendocrine carcinomas (NECs). The site of origin of the NENs is linked to the intrinsic tumour biology and is predictive of the disease course. It is understood that NENs demonstrate significant biologic heterogeneity which ultimately translates to widely varying clinical presentations, disease course and prognosis. Thus, significant emphasis is laid on the pre-therapy evaluation of markers that can help predict tumour behavior and dynamically monitors the response during and after treatment. Most well-differentiated NENs express somatostatin receptors (SSTRs) which make them appropriate for peptide receptor radionuclide therapy (PRRT). However, the treatment outcomes of PRRT depend heavily on the adequacy of patient selection by molecular imaging phenotyping not only utilizing pre-treatment SSTR PET but 18F-Fluorodeoxyglucose (18F-FDG) PET to provide insights into the intra- or inter-tumoural heterogeneity of the metastatic disease. Molecular imaging phenotyping may go beyond patient selection and provide useful information during and post-treatment for monitoring of temporal heterogeneity of the disease and dynamically risk-stratify patients. In addition, advances in the understanding of genomic-phenotypic classifications of pheochromocytomas and paragangliomas led to an archetypical example in precision medicine by utilizing molecular imaging phenotyping to guide radioligand therapy. Novel non-SSTR based peptide receptors have also been explored diagnostically and therapeutically to overcome the tumour heterogeneity. In this paper, we review the current molecular imaging modalities that are being utilized for the characterization of the NENs with special emphasis on their role in patient selection for radioligand therapy.
Collapse
|
21
|
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
|
22
|
Galgano SJ, Iravani A, Bodei L, El-Haddad G, Hofman MS, Kong G. Imaging of Neuroendocrine Neoplasms: Monitoring Treatment Response- AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2022; 218:767-780. [PMID: 34985313 DOI: 10.2214/ajr.21.27159] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Neuroendocrine neoplasms (NENs) encompass a broad spectrum of tumors throughout the body and range in biologic behavior from indolent to aggressive. Consequently, a wide spectrum of treatment options are available for NENs, including observation, somatostatin analogues, targeted therapy, chemotherapy, surgical resection, liver-directed therapy (embolization and ablation), and peptide receptor radionuclide therapy. Given the wide variety of tumor behaviors and treatments, precise criteria for treatment response in NENs are lacking. Though conventional anatomic imaging with CT and MRI remains important for NEN response assessment, the use of somatostatin receptor (SSR) PET is increasing and often provides synergistic and complementary information. Additionally, in certain clinical scenarios, a particular imaging strategy may prove superior or inferior to others for the detection of metastatic disease and evaluation of therapy response. A strong need exists to further define appropriate and standardized assessment criteria for tumor response and progression in NEN. This article presents the strengths and weaknesses of individual imaging modalities for evaluating NEN therapy response, including conventional anatomic imaging, SSR PET, FDG PET, dual-tracer PET, and PET/MRI. Ongoing challenges and unmet needs in the use of imaging for NEN response evaluation are explored.
Collapse
Affiliation(s)
- Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, 619 19th St S, JT N325, Birmingham, AL 35249
| | - Amir Iravani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO
| | - Lisa Bodei
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ghassan El-Haddad
- Department of Diagnostic Imaging and Interventional Radiology, Moffitt Cancer Center, Tampa, FL
| | - Michael S Hofman
- Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Grace Kong
- Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
23
|
Mejia A, Vivian E, Nwogu C, Shah J, Longoria R, Vo A, Shahin I, Verma J, Bageac A. Peptide receptor radionuclide therapy implementation and results in a predominantly gastrointestinal neuroendocrine tumor population: A two-year experience in a nonuniversity setting. Medicine (Baltimore) 2022; 101:e28970. [PMID: 35244064 PMCID: PMC8896579 DOI: 10.1097/md.0000000000028970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/10/2022] [Indexed: 01/04/2023] Open
Abstract
Neuroendocrine tumors (NETs) are rare, but the incidence and prevalence of NETs are increasing in the United States. While surgery is the preferred treatment for NETs, it is not a viable option for metastatic disease. Lutathera (177Lu-DOTATATE) is approved by the United States Food and Drug Administration and the European Medicines Agency for the treatment of gastroenteropancreatic (GEP)-NETs in adults. There is limited information on GEP-NET treatment responses to Lutathera.Our institution launched a peptide receptor radionuclide therapy (PRRT) service line using Lutathera with involvement from a multidisciplinary team and complete collaboration between hospital administration and clinical providers. A prospective registry study was also established in order to collect patient demographics and clinical data regarding the treatment of GEP primary NETs with Lutathera.Between August 2018 and July 2020, 35 GEP-NET patients were treated with Lutathera, of which 65.71% received 4 complete cycles and 25.71% received 3 cycles; 5.71% and 2.86% received 2 and 1 cycles of PRRT, respectively. Most adverse events during the course of our study were low grade using the common terminology criteria for adverse events system. Of the patients who completed all 4 cycles: 22% showed partial response to Lutathera, 44% showed stable disease, and 13% showed disease progression based on a qualitative assessment of positron emission tomography/computed tomography imaging.From our experience, Lutathera was well tolerated in patients with GEP-NET. Additional studies are needed to examine long-term clinical and patient-reported outcomes associated with GEP-NET treatment as well as financial considerations for hospitals embarking on a PRRT program.
Collapse
Affiliation(s)
- Alejandro Mejia
- The Liver Institute, Methodist Dallas Medical Center, Dallas, TX
| | - Elaina Vivian
- Methodist Digestive Institute, Methodist Dallas Medical Center, Dallas, TX
| | - Christiana Nwogu
- Methodist Digestive Institute, Methodist Dallas Medical Center, Dallas, TX
| | - Jimmy Shah
- Methodist Digestive Institute, Methodist Dallas Medical Center, Dallas, TX
| | - Raquel Longoria
- Cancer Program Administration, Methodist Dallas Medical Center, Dallas, TX
| | - Allison Vo
- Methodist Digestive Institute, Methodist Dallas Medical Center, Dallas, TX
- Cancer Program Administration, Methodist Dallas Medical Center, Dallas, TX
| | - Islam Shahin
- Radiology Associates of North Texas, Fort Worth, TX
| | - Jonathan Verma
- Texas Oncology – Methodist Dallas Medical Center, Dallas, TX
| | | |
Collapse
|
24
|
Dromain C, Vullierme M, Hicks RJ, Prasad V, O’Toole D, de Herder WW, Pavel M, Faggiano A, Kos‐Kudla B, Öberg K, Krejs GJ, Grande E, Niederle B, Sundin A. ENETS standardized (synoptic) reporting for radiological imaging in neuroendocrine tumours. J Neuroendocrinol 2022; 34:e13044. [PMID: 34693574 PMCID: PMC9286653 DOI: 10.1111/jne.13044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/06/2021] [Indexed: 12/05/2022]
Abstract
This expert consensus document represents an initiative by the European Neuroendocrine Tumor Society (ENETS) to provide guidance for synoptic reporting of radiological examinations critical to the diagnosis, grading, staging and treatment of neuroendocrine neoplasms (NENs). Template drafts for initial tumor staging and follow-up by computed tomography (CT) and magnetic resonance imaging (MRI) were established, based on existing institutional and organisational reporting templates relevant for NEN imaging, and applying the RadLex lexicon of radiological information (Radiological Society of North America), for consistency regarding the radiological terms. During the ENETS Scientific Advisory Board meeting 2018, the template drafts were subject to iterative interdisciplinary discussions among experts in imaging, surgery, gastroenterology, oncology and pathology. Members of the imaging group stated a strong preference for a combination of limited and standardised options by way of drop-down menus. Separate templates were produced for the initial work-up and for follow-up, respectively. To provide a detailed description of the radiological findings of the primary tumor and its local extension and spread, different templates were developed for bronchial, pancreatic and gastrointestinal NENs for CT and MRI, respectively. Each template was structured in 10 sections: clinical details, comparative imaging modality, acquisition technique, primary tumor findings, regional lymph node metastases, distant metastases, TNM classification, reference lesions according to RECIST 1.1, additional findings and conclusion. Two templates were developed for follow-up, for CT and MRI, respectively, and were specifically focused on assessment of therapy response. These included a qualitative response assessment, such as decrease of vascularisation and presence of necrosis, and a quantitative assessment according to RECIST 1.1 and the modified RECIST (mRECIST) for assessing tumor response following transarterial chemoembolisation.
Collapse
Affiliation(s)
- Clarisse Dromain
- Department of RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Marie‐Pierre Vullierme
- Department of RadiologyHôpital Beaujon – Hôpitaux Universitaires Paris Nord Val de SeineUniversité de ParisParisFrance
| | - Rodney J. Hicks
- Neuroendocrine ServiceThe Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVICAustralia
| | - Vikas Prasad
- Department of Nuclear MedicineUniversity UlmUlmGermany
| | - Dermot O’Toole
- St. James's and St. Vincent's University Hospitals & Trinity College DublinDublinRepublic of Ireland
| | - Wouter W. de Herder
- Department of Internal MedicineSection of EndocrinologyErasmus MCRotterdamThe Netherlands
| | - Marianne Pavel
- Department of Medicine 1Universitätsklinikum ErlangenErlangenGermany
| | - Antongiulio Faggiano
- Department of Clinical and Molecular MedicineSapienza University of Rome, Sant'Andrea HospitalRomeItaly
| | - Beata Kos‐Kudla
- Department of Endocrinology and Neuroendocrine TumorsMedical University of SilesiaKatowicePoland
| | - Kjell Öberg
- Department of Endocrine OncologyUniversity Hospital UppsalaUppsalaSweden
| | - Guenter J. Krejs
- Division of Gastroenterology and Hepatology, Department of Internal MedicineMedical University of GrazGrazAustria
| | - Enrique Grande
- Department of Medical OncologyMD Anderson Cancer Center MadridMadridSpain
| | - Bruno Niederle
- Department of General SurgeryMedical University of ViennaViennaAustria
| | - Anders Sundin
- Department of Surgical SciencesRadiology & Molecular Imaging, Uppsala University HospitalUppsala UniversityUppsalaSweden
| |
Collapse
|
25
|
van Treijen MJC, Schoevers JMH, Heeres BC, van der Zee D, Maas M, Valk GD, Tesselaar MET. Defining disease status in gastroenteropancreatic neuroendocrine tumors: Choi-criteria or RECIST? Abdom Radiol (NY) 2022; 47:1071-1081. [PMID: 34989825 DOI: 10.1007/s00261-021-03393-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Adequate monitoring of changes in tumor load is fundamental for the assessment of the course of disease and response to treatment. There is an ongoing debate on the utility of RECIST v1.1 in gastroenteropancreatic neuroendocrine tumors (GEP-NETs). METHODS In this retrospective real-life cohort study, Choi-criteria were compared with RECIST v1.1. The agreement between both criteria and the association with survival endpoints were evaluated. RESULTS Seventy-five patients were included with a median follow-up of 35 months (range 8-53). Median progression-free survival (mPFS) according to RECIST v1.1 was 15 months (range 2-50) compared to 14 months (range 2-50) in Choi. According to RECIST, 33 (44%) patients were classified as having stable disease (SD), 40 (53%) as progressive disease (PD) and two (3%) patients as partial response (PR), compared to 9 (12%) patients classified as SD, 50 (67%) as PD and 16 (21%) as PR according to Choi-criteria. Overall concordance between the criteria was moderate (Cohen's Kappa = 0.408, p < 0.001) and agreement varied between 57 and 69% at each consecutive scan (p < 0.001). Survival analysis showed significant differences in overall survival (OS) for RECIST v1.1 categories PD and non-PD (log-rank p = 0.02), however, in Choi no significant differences in OS were found (p = 0.27). CONCLUSION RECIST v1.1 had a better clinical utility and prognostic value compared to Choi-criteria. Still, RECIST were also not sufficient to adequately predict OS. This outlines the need for new tools that provides accurate information on the disease course and treatment response to support precise prognostication in patients with GEP-NETs.
Collapse
Affiliation(s)
- M J C van Treijen
- Department of Endocrine Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands.
- The Netherlands Cancer Institute/University Medical Center Utrecht Center for Neuroendocrine Tumors, ENETs Center of Excellence, Amsterdam/Utrecht, The Netherlands.
| | - J M H Schoevers
- Department of Endocrine Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
| | - B C Heeres
- The Netherlands Cancer Institute/University Medical Center Utrecht Center for Neuroendocrine Tumors, ENETs Center of Excellence, Amsterdam/Utrecht, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - D van der Zee
- Department of Radiology, Bernhoven Hospital, Uden, The Netherlands
| | - M Maas
- The Netherlands Cancer Institute/University Medical Center Utrecht Center for Neuroendocrine Tumors, ENETs Center of Excellence, Amsterdam/Utrecht, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - G D Valk
- Department of Endocrine Oncology, University Medical Center Utrecht Cancer Center, Heidelberglaan 100, 3584CX, Utrecht, The Netherlands
- The Netherlands Cancer Institute/University Medical Center Utrecht Center for Neuroendocrine Tumors, ENETs Center of Excellence, Amsterdam/Utrecht, The Netherlands
| | - M E T Tesselaar
- The Netherlands Cancer Institute/University Medical Center Utrecht Center for Neuroendocrine Tumors, ENETs Center of Excellence, Amsterdam/Utrecht, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Lee L, Ramos-Alvarez I, Jensen RT. Predictive Factors for Resistant Disease with Medical/Radiologic/Liver-Directed Anti-Tumor Treatments in Patients with Advanced Pancreatic Neuroendocrine Neoplasms: Recent Advances and Controversies. Cancers (Basel) 2022; 14:1250. [PMID: 35267558 PMCID: PMC8909561 DOI: 10.3390/cancers14051250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose: Recent advances in the diagnosis, management and nonsurgical treatment of patients with advanced pancreatic neuroendocrine neoplasms (panNENs) have led to an emerging need for sensitive and useful prognostic factors for predicting responses/survival. Areas covered: The predictive value of a number of reported prognostic factors including clinically-related factors (clinical/laboratory/imaging/treatment-related factors), pathological factors (histological/classification/grading), and molecular factors, on therapeutic outcomes of anti-tumor medical therapies with molecular targeting agents (everolimus/sunitinib/somatostatin analogues), chemotherapy, radiological therapy with peptide receptor radionuclide therapy, or liver-directed therapies (embolization/chemoembolization/radio-embolization (SIRTs)) are reviewed. Recent findings in each of these areas, as well as remaining controversies and uncertainties, are discussed in detail, particularly from the viewpoint of treatment sequencing. Conclusions: The recent increase in the number of available therapeutic agents for the nonsurgical treatment of patients with advanced panNENs have raised the importance of prognostic factors predictive for therapeutic outcomes of each treatment option. The establishment of sensitive and useful prognostic markers will have a significant impact on optimal treatment selection, as well as in tailoring the therapeutic sequence, and for maximizing the survival benefit of each individual patient. In the paper, the progress in this area, as well as the controversies/uncertainties, are reviewed.
Collapse
Affiliation(s)
- Lingaku Lee
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
- National Kyushu Cancer Center, Department of Hepato-Biliary-Pancreatology, Fukuoka 811-1395, Japan
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
| | - Robert T. Jensen
- Digestive Diseases Branch, NIDDK, NIH, Bethesda, MD 20892-1804, USA; (L.L.); (I.R.-A.)
| |
Collapse
|
27
|
Terapia con péptidos radiomarcados con [177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Peptide Receptor Radionuclide Therapy Targeting the Somatostatin Receptor: Basic Principles, Clinical Applications and Optimization Strategies. Cancers (Basel) 2021; 14:cancers14010129. [PMID: 35008293 PMCID: PMC8749814 DOI: 10.3390/cancers14010129] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/13/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Peptide receptor radionuclide therapy (PRRT) is a systemic treatment consisting of the administration of a tumor-targeting radiopharmaceutical into the circulation of a patient. The radiopharmaceutical will bind to a specific peptide receptor leading to tumor-specific binding and retention. This will subsequently cause lethal DNA damage to the tumor cell. The only target that is currently used in widespread clinical practice is the somatostatin receptor, which is overexpressed on a range of tumor cells, including neuroendocrine tumors and neural-crest derived tumors. Academia played an important role in the development of PRRT, which has led to heterogeneous literature over the last two decades, as no standard radiopharmaceutical or regimen has been available for a long time. This review focuses on the basic principles and clinical applications of PRRT, and discusses several PRRT-optimization strategies. Abstract Peptide receptor radionuclide therapy (PRRT) consists of the administration of a tumor-targeting radiopharmaceutical into the circulation of a patient. The radiopharmaceutical will bind to a specific peptide receptor leading to tumor-specific binding and retention. The only target that is currently used in clinical practice is the somatostatin receptor (SSTR), which is overexpressed on a range of tumor cells, including neuroendocrine tumors and neural-crest derived tumors. Academia played an important role in the development of PRRT, which has led to heterogeneous literature over the last two decades, as no standard radiopharmaceutical or regimen has been available for a long time. This review provides a summary of the treatment efficacy (e.g., response rates and symptom-relief), impact on patient outcome and toxicity profile of PRRT performed with different generations of SSTR-targeting radiopharmaceuticals, including the landmark randomized-controlled trial NETTER-1. In addition, multiple optimization strategies for PRRT are discussed, i.e., the dose–effect concept, dosimetry, combination therapies (i.e., tandem/duo PRRT, chemoPRRT, targeted molecular therapy, somatostatin analogues and radiosensitizers), new radiopharmaceuticals (i.e., SSTR-antagonists, Evans-blue containing vector molecules and alpha-emitters), administration route (intra-arterial versus intravenous) and response prediction via molecular testing or imaging. The evolution and continuous refinement of PRRT resulted in many lessons for the future development of radionuclide therapy aimed at other targets and tumor types.
Collapse
|
29
|
Prado-Wohlwend S, Bernal-Vergara JC, Utrera-Costero A, Cañón-Sánchez JR, Agudelo-Cifuentes M, Bello-Arques P. Peptide receptor radionuclide therapy with [ 177Lu]Lu-DOTA-TATE. Rev Esp Med Nucl Imagen Mol 2021; 41:55-65. [PMID: 34920969 DOI: 10.1016/j.remnie.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022]
Abstract
This continuing education aims to present in a clear and easy-to-understand way, the biology of neuroendocrine tumors (NETs), the characteristics of somatostatin receptors, the selection of patients for radiolabelled peptide therapy (PRRT), the inclusion criteria to benefit from treatment with the minimum possible adverse effects, the administration protocol, follow-up and response evaluation. The functional imaging studies necessary to explore the biology of the tumor and to individualize the treatment are also carried out, and constitute the cornerstone for the development of teragnosis. Clinical trials are being developed to better define the position of PRRT within the broad therapeutic options, and among the future perspectives, there are several lines of research to improve the objective response rate and survival with PRRT, focused on the development of new agonists and somatostatin receptor antagonists, new radionuclides and radiosensitizing combination therapies. In conclusion, PRRT is a great therapeutic, well-tolerated and safe tool with generally mild and self-limited acute side effects, that must be sequenced at the best moment of the evolution of the disease of patients with NET. Candidate patients for PRRT should always be evaluated by a multidisciplinary clinical committee.
Collapse
Affiliation(s)
- S Prado-Wohlwend
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain.
| | - J C Bernal-Vergara
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - A Utrera-Costero
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - J R Cañón-Sánchez
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M Agudelo-Cifuentes
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - P Bello-Arques
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | -
- Servicio de Medicina Nuclear, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
30
|
Vullierme MP, Ruszniewski P, de Mestier L. Are recist criteria adequate in assessing the response to therapy in metastatic NEN? Rev Endocr Metab Disord 2021; 22:637-645. [PMID: 33871762 DOI: 10.1007/s11154-021-09645-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Response to therapy criteria, known as RECIST (Response Evaluation Criteria in Solid Tumours), are widely used to evaluate neuroendocrine tumours (NET) metastatic to the liver, under treatment. RECIST criteria does not take in account many various distinct features such as tumour growth, secretory capacity and anatomical localisation with wide variation in clinical and biological presentation of different NETs. Key features of RECIST includes definitions of the minimal size of measurable lesions, instructions on how many lesions to measure and follow, and the use of unidimensional, rather than bidimensional, measures for overall evaluation of tumour burden. These measures are currently done with computed tomography (CT) or Magnetic Resonance Imaging (MRI). RECIST criteria are accurate in assessing tumour progression but sometimes inaccurate in assessing tumour response after locoregional therapy or under molecular targeted therapy, tumour vessels being part of the target of such treatments. There is poor correlation between a so called tumour necrosis and conventional methods of response assessment, which poses questions of how best to quantify efficacy of these targeted therapies. Variations in tumour density with computed tomography (CT) could theoretically be associated with tumour necrosis. This hypothesis has been studied proposing alternative CT criteria of response evaluation in metastatic digestive NET treated with targeted therapy. If preliminary results upon the poor relationship between density measured with CT (derived from CHOI criteria) evolution curves at CT and PFS are confirmed by further studies, showing that the correlation between density changing and response to non-targeted treatment is weak, the use of contrast injection, will probably be not mandatory to enable appropriate evaluation.
Collapse
Affiliation(s)
- Marie-Pierre Vullierme
- Department of Radiology, University Hospitals Paris-Nord Val-de-Seine, AP-HP, Beaujon, 92110, Clichy, France.
- Université Paris Diderot-Paris 7, University of Paris, 75018, Clichy, France.
| | - Philippe Ruszniewski
- Department of Pancreatology, University Hospitals Paris-Nord Val-de-Seine, AP-HP, Beaujon, 92110, Clichy, France
- Université Paris Diderot-Paris 7, University of Paris, 75018, Clichy, France
| | - Louis de Mestier
- Department of Pancreatology, University Hospitals Paris-Nord Val-de-Seine, AP-HP, Beaujon, 92110, Clichy, France
- Université Paris Diderot-Paris 7, University of Paris, 75018, Clichy, France
| |
Collapse
|
31
|
68Ga-DOTATATE PET/CT and MRI with Diffusion-Weighted Imaging (DWI) in Short- and Long-Term Assessment of Tumor Response of Neuroendocrine Liver Metastases (NELM) Following Transarterial Radioembolization (TARE). Cancers (Basel) 2021; 13:cancers13174321. [PMID: 34503131 PMCID: PMC8431353 DOI: 10.3390/cancers13174321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023] Open
Abstract
Simple Summary TARE with 90Yttrium has become a valuable treatment option for patients with unresectable NELMs. However, early evaluation of therapy response remains challenging as size-based response assessments (such as RECIST) are known to be limited, especially in slow-growing tumors. Alternatives such as quantitative evaluation of SUV of 68Ga-DOTATATE PET/CT and ADC of DWI-MRI have not been compared so far. We found that early percentage changes in SUV tumor-to-organ ratios on first follow-up after TARE could predict longer HPFS in patients with NELM and were superior to ΔSUVmax/SUVmean alone or to ΔADC. Abstract The aim of this study was to evaluate the role of SUV and ADC in assessing early response in patients with NELM following TARE. Thirty-two patients with pre- and postinterventional MRI with DWI and 68Ga-DOTATATE PET/CT were included. ADC and SUV of three target lesions and of tumor-free spleen and liver tissue were determined on baseline and first follow-up imaging, and tumor to spleen (T/S) and tumor to liver (T/L) ratios were calculated. Response was assessed by RECIST 1.1 and mRECIST on first follow-up, and long-term response was defined as hepatic progression-free survival (HPFS) over 6, 12, and <24 months. In responders, intralesional ADC values increased and SUV decreased significantly regardless of standard of reference for response assessment (mRECIST/RECIST/HPFS > 6/12/24 m). Using ROC analysis, ΔSUV T/S ratio (max/max) and ΔSUV T/L ratio (max/mean) were found to be the best and most robust metrics to correlate with longer HPFS and were superior to ΔADC. ΔT/S ratio (max/max) < 23% was identified as an optimal cut-off to discriminate patients with longer HPFS (30.2 m vs. 13.4 m; p = 0.0002). In conclusion, early percentage changes in SUV tumor-to-organ ratios on first follow-up seem to represent a prognostic marker for longer HPFS and may help in assessing therapeutic strategies.
Collapse
|
32
|
Ferdinandus J, Fendler WP, Morigi JJ, Fanti S. Theranostics in oncology: What radiologists want to know. Eur J Radiol 2021; 142:109875. [PMID: 34391057 DOI: 10.1016/j.ejrad.2021.109875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/15/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022]
Abstract
Combination of radioligand imaging and therapy, so called radiotheranostics, is a novel tool of precision oncology with proven clinical value. In-depth knowledge of functional imaging nuances is critically needed for precise prognostication and guidance of management. Here, we review theranostic applications with up to Phase III type evidence for outcome improvement: Imaging and therapy of neuroendocrine neoplasms (NEN) exploiting high levels of somatostatin receptor (SSTR) expression and radiotheranostics of prostate cancer targeting the prostate specific membrane antigen (PSMA). This narrative review focusses on these two applications and elucidates patient selection and response assessment by radioligand scintigraphy and/or positron emission tomography. Furthermore, we provide a brief outlook on future applications for novel targets outside of NEN and prostate cancer.
Collapse
Affiliation(s)
- Justin Ferdinandus
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Wolfgang Peter Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Joshua James Morigi
- PET/CT Unit, Department of Medical Imaging, Royal Darwin Hospital, Darwin, Australia.
| | - Stefano Fanti
- Nuclear Medicine Division, Policlinico S Orsola, University of Bologna, Bologna, Italy
| |
Collapse
|
33
|
Abstract
This article summarizes the role of PET imaging for detection, characterization, and theranostic/therapy planning for neuroendocrine tumors. Topics in this article span overall imaging accuracy with mostly 68Ga-DOTA-peptide imaging as well as basic principles of individualized dosimetry. There is also some discussion around further specialized approaches in dosimetry in theranostics. In addition, an overview of the literature on functional imaging in neuroendocrine tumors and the current understanding of imaging-derived clinical outcome prediction are presented.
Collapse
Affiliation(s)
- Rebecca K S Wong
- Radiation Medicine Program, Princess Margaret Cancer Center, University Health Network, 610 University Ave, Toronto, ON M5G 2M9, Canada; Department of Radiation Oncology Temerty Faculty of Medicine, University of Toronto, 149 College Street, Suite 504, Toronto, ON M5T 1P5, Canada
| | - Ur Metser
- Joint Department Medical Imaging, University Health Network, Mount Sinai Hospital & Women's College Hospital, University of Toronto, 610 University Ave, Suite 3-920, Toronto, ON M5G 2M9, Canada; Department of Medical Imaging, University of Toronto, 263 McCaul Street, 4th Floor, Toronto, ON M5T 1W7, Canada
| | - Patrick Veit-Haibach
- Joint Department Medical Imaging, University Health Network, Mount Sinai Hospital & Women's College Hospital, University of Toronto, 610 University Ave, Suite 3-920, Toronto, ON M5G 2M9, Canada; Department of Medical Imaging, University of Toronto, 263 McCaul Street, 4th Floor, Toronto, ON M5T 1W7, Canada.
| |
Collapse
|
34
|
Roll W, Weckesser M, Seifert R, Bodei L, Rahbar K. Imaging and liquid biopsy in the prediction and evaluation of response to PRRT in neuroendocrine tumors: implications for patient management. Eur J Nucl Med Mol Imaging 2021; 48:4016-4027. [PMID: 33903926 PMCID: PMC8484222 DOI: 10.1007/s00259-021-05359-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022]
Abstract
Purpose The aim of this narrative review is to give an overview on current and emerging imaging methods and liquid biopsy for prediction and evaluation of response to PRRT. Current limitations and new perspectives, including artificial intelligence, are discussed. Methods A literature review of PubMed/Medline was performed with representative keywords. The search included articles published online through August 31, 2020. All searches were restricted to English language manuscripts. Results Peptide radio receptor therapy (PRRT) is a prospectively evaluated and approved therapy option in neuroendocrine tumors (NETs). Different ligands targeting the somatostatin receptor (SSTR) are used as theranostic pairs for imaging NET and for PRRT. Response assessment in prospective trials often relies on the morphological RECIST 1.1 criteria, based on lesion size in CT or MRI. The role of SSTR-PET and quantitative uptake parameters and volumetric data is still not defined. Monoanalyte tumor marker chromogranin A has a limited value for response assessment after PRRT. New emerging liquid biopsy techniques are offering prediction of response to PRRT and prognostic value. Conclusions New response criteria for NET patients undergoing PRRT will comprise multiparametric hybrid imaging and blood-based multianalyte markers. This represents tumor biology and heterogeneity.
Collapse
Affiliation(s)
- Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.,West German Cancer Center, Muenster and Essen, Essen, Germany.,Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Lisa Bodei
- Department of Nuclear Medicine, Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany. .,West German Cancer Center, Muenster and Essen, Essen, Germany.
| |
Collapse
|
35
|
Das S, Du L, Schad A, Jain S, Jessop A, Shah C, Eisner D, Cardin D, Ciombor K, Goff L, Bradshaw M, Delbeke D, Sandler M, Berlin J. A clinical score for neuroendocrine tumor patients under consideration for Lu-177-DOTATATE therapy. Endocr Relat Cancer 2021; 28:203-212. [PMID: 33608484 PMCID: PMC8026653 DOI: 10.1530/erc-20-0482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 01/01/2023]
Abstract
We developed a clinical score (CS) at Vanderbilt Ingram Cancer Center (VICC) that we hoped would predict outcomes for patients with progressive well-differentiated neuroendocrine tumors (NETs) receiving therapy with Lutetium-177 (177Lu)-DOTATATE. Patients under consideration for 177Lu-DOTATATE between March 1, 2016 and March 17, 2020 at VICC were assigned a CS prospectively. The CS included 5 categories: available treatments for tumor type outside of 177Lu-DOTATATE, prior systemic treatments, patient symptoms, tumor burden in critical organs and presence of peritoneal carcinomatosis. The primary outcome of the analysis was progression-free survival (PFS). To evaluate the effect of the CS on PFS, a multivariable Cox regression analysis was performed adjusting for tumor grade, primary tumor location, and the interaction between 177Lu-DOTATATE doses received (zero, 1-2, 3-4) and CS. A total of 91 patients and 31 patients received 3-4 doses and zero doses of 177Lu-DOTATATE, respectively. On multivariable analysis, in patients treated with 3-4 doses of 177Lu-DOTATATE, for each 1-point increase in CS, the estimated hazard ratio (HR) for PFS was 2.0 (95% CI 1.61-2.48). On multivariable analysis, in patients who received zero doses of 177Lu-DOTATATE, for each 1-point increase in CS, the estimated HR for PFS was 1.22 (95% CI 0.91-1.65). Among patients treated with 3-4 doses of 177Lu-DOTATATE, those with lower CS experienced improved PFS with the treatment compared to patients with higher CS. This PFS difference, based upon CS, was not observed in patients who did not receive 177Lu-DOTATATE, suggesting the predictive utility of the score.
Collapse
Affiliation(s)
- Satya Das
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| | - Liping Du
- Vanderbilt University of Medical Center, Department of Biostatistics, Nashville, TN, USA
| | - Aimee Schad
- Rush Medical Center, Department of Medicine, Chicago, IL, USA
| | - Shikha Jain
- University of Illinois Chicago, Department of Medicine, Chicago, IL, USA
| | - Aaron Jessop
- Vanderbilt University Medical Center, Department of Radiology, Nashville, TN, USA
| | - Chirayu Shah
- Vanderbilt University Medical Center, Department of Radiology, Nashville, TN, USA
| | - David Eisner
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| | - Dana Cardin
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| | - Kristen Ciombor
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| | - Laura Goff
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| | - Marques Bradshaw
- Vanderbilt University Medical Center, Department of Radiology, Nashville, TN, USA
| | - Dominique Delbeke
- Vanderbilt University Medical Center, Department of Radiology, Nashville, TN, USA
| | - Martin Sandler
- Vanderbilt University Medical Center, Department of Radiology, Nashville, TN, USA
| | - Jordan Berlin
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology Oncology, Nashville, TN, USA
| |
Collapse
|
36
|
Liberini V, Huellner MW, Grimaldi S, Finessi M, Thuillier P, Muni A, Pellerito RE, Papotti MG, Piovesan A, Arvat E, Deandreis D. The Challenge of Evaluating Response to Peptide Receptor Radionuclide Therapy in Gastroenteropancreatic Neuroendocrine Tumors: The Present and the Future. Diagnostics (Basel) 2020; 10:E1083. [PMID: 33322819 PMCID: PMC7763988 DOI: 10.3390/diagnostics10121083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The NETTER-1 study has proven peptide receptor radionuclide therapy (PRRT) to be one of the most effective therapeutic options for metastatic neuroendocrine tumors (NETs), improving progression-free survival and overall survival. However, PRRT response assessment is challenging and no consensus on methods and timing has yet been reached among experts in the field. This issue is owed to the suboptimal sensitivity and specificity of clinical biomarkers, limitations of morphological response criteria in slowly growing tumors and necrotic changes after therapy, a lack of standardized parameters and timing of functional imaging and the heterogeneity of PRRT protocols in the literature. The aim of this article is to review the most relevant current approaches for PRRT efficacy prediction and response assessment criteria in order to provide an overview of suitable tools for safe and efficacious PRRT.
Collapse
Affiliation(s)
- Virginia Liberini
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Serena Grimaldi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Monica Finessi
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| | - Philippe Thuillier
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
- Department of Endocrinology, University Hospital of Brest, 29200 Brest, France
| | - Alfredo Muni
- Department of Nuclear Medicine, S.S. Biagio e Antonio e C. Arrigo Hospital, 15121 Alessandria, Italy;
| | | | - Mauro G. Papotti
- Pathology Unit, City of Health and Science University Hospital, 10126 Turin, Italy;
- Department of Oncology, University of Turin at Molinette Hospital, 10126 Turin, Italy
| | - Alessandro Piovesan
- Department of Endocrinology, A. O. U. Città della Salute della Scienza of Turin, 10126 Turin, Italy;
| | - Emanuela Arvat
- Oncological Endocrinology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Désirée Deandreis
- Nuclear Medicine Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (S.G.); (M.F.); (P.T.); (D.D.)
| |
Collapse
|
37
|
Calabrò D, Argalia G, Ambrosini V. Role of PET/CT and Therapy Management of Pancreatic Neuroendocrine Tumors. Diagnostics (Basel) 2020; 10:E1059. [PMID: 33297381 PMCID: PMC7762240 DOI: 10.3390/diagnostics10121059] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are heterogeneous neoplasms with neuroendocrine differentiation that show peculiar clinical and histomorphological features, with variable prognosis. In recent years, advances in knowledge regarding the pathophysiology and heterogeneous clinical presentation, as well as the availability of different diagnostic procedures for panNEN diagnosis and novel therapeutic options for patient clinical management, has led to the recognition of the need for an active multidisciplinary discussion for optimal patient care. Molecular imaging with positron emission tomography/computed tomography (PET/CT) has become indispensable for the management of panNENs. Several PET radiopharmaceuticals can be used to characterize either panNEN receptor expression or metabolism. The aim of this review is to offer an overview of all the currently used radiopharmaceuticals and of the new upcoming tracers for pancreatic neuroendocrine tumors (panNETs), and their clinical impact on therapy management. [68Ga]Ga-DOTA-peptide PET/CT (SSA-PET/CT) has high sensitivity, specificity, and accuracy and is recommended for the staging and restaging of any non-insulinoma well-differentiated panNEN cases to carry out detection of unknown primary tumor sites or early relapse and for evaluation of in vivo somatostatin receptors expression (SRE) to select patient candidates for peptide receptor radiometabolic treatment (PRRT) with 90Y or 177Lu and/or cold analogs. SSA-PET/CT also has a strong impact on clinical management, leading to a change in treatment in approximately a third of the cases. Its role for treatment response assessment is still under debate due to the lack of standardized criteria, even though some semiquantitative parameters seem to be able to predict response. [18F]FDG PET/CT generally shows low sensitivity in small growing and well-differentiated neuroendocrine tumors (NET; G1 and G2), while it is of utmost importance in the evaluation and management of high-grade NENs and also provides important prognostic information. When positive, [18F]FDG PET/CT impacts therapeutical management, indicating the need for a more aggressive treatment regime. Although FDG positivity does not exclude the patient from PRRT, several studies have demonstrated that it is certainly useful to predict response, even in this setting. The role of [18F]FDOPA for the study of panNET is limited by physiological uptake in the pancreas and is therefore not recommended. Moreover, it provides no information on SRE that has crucial clinical management relevance. Early acquisition of the abdomen and premedication with carbidopa may be useful to increase the accuracy, but further studies are needed to clarify its utility. GLP-1R agonists, such as exendin-4, are particularly useful for benign insulinoma detection, but their accuracy decreases in the case of malignant insulinomas. Being a whole-body imaging technique, exendin-PET/CT gives important preoperative information on tumor size and localization, which is fundamental for surgical planning as resection (enucleation of the lesion or partial pancreatic resection) is the only curative treatment. New upcoming tracers are under study, such as promising SSTR antagonists, which show a favorable biodistribution and higher tumor-to-background ratio that increases tumor detection, especially in the liver. [68Ga]pentixafor, an in vivo marker of CXCR4 expression associated with the behavior of more aggressive tumors, seems to only play a limited role in detecting well-differentiated NET since there is an inverse expression of SSTR2 and CXCR4 in G1 to G3 NETs with an elevation in CXCR4 and a decrease in SSTR2 expression with increasing grade. Other tracers, such as [68Ga]Ga-PSMA, [68Ga]Ga-DATA-TOC, [18F]SiTATE, and [18F]AlF-OC, are also under investigation.
Collapse
Affiliation(s)
- Diletta Calabrò
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| | - Valentina Ambrosini
- Department of Nuclear Medicine, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy; (G.A.); (V.A.)
- Department of Nuclear Medicine, DIMES University of Bologna, 40138 Bologna, Italy
| |
Collapse
|