1
|
Yamaguchi N, Wei JJ, Isomoto H. Clinical application of targeted α-emitter therapy in gastroenteropancreatic neuroendocrine neoplasms. J Gastroenterol 2025:10.1007/s00535-025-02241-z. [PMID: 40220045 DOI: 10.1007/s00535-025-02241-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 04/14/2025]
Abstract
Effective therapeutic strategies for advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remain challenging, including a lack of response to therapy and post-treatment relapse. The rapid development of targeted radionuclide therapy (TRT) offers promising data for patients with somatostatin receptor (SSTR)-expressing tumors. This approach exhibits more advantages than somatostatin analog (SSA) therapy, which is primarily effective for well-differentiated and slow-growing GEP-NENs. Fortunately, some clinical studies on peptide receptor radionuclide therapy (PRRT) labeled with α-emitting radionuclides for GEP-NENs patients showed effective results for those with more advanced GEP-NENs, or those with malignant metastasis. For the improvement of clinical efficacy and the decline in the incidence of treatment-related relapse, recent progress in developing novel techniques and effective disease management strategies for optimal targeting has led to the emergence of targeted alpha therapy (TAT) in GEP-NENs patients. For instance, labeled technology and combination therapy could contribute to significantly improved long-term outcomes. However, the exact dosimetry for precision oncology, the shortage of radionuclides, and the stability of disease control are still under careful consideration. More high-quality, large-scale prospective studies are essential for obtaining valuable evidence on challenging problems and for further exploration.
Collapse
Affiliation(s)
- Naoyuki Yamaguchi
- Department of Endoscopy, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Nagasaki, 852-8501, Japan
| | - Jing-Jing Wei
- Department of Endoscopy, the First Affiliated Hospital of Fujian Medical University, Cha Zhong Road No.20, Tai Jiang District, Fuzhou, 350004, Fujian, China.
- Department of Endoscopy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan.
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan
| |
Collapse
|
2
|
Salles-Silva E, de Castro PL, Ambrozino LC, de Araújo ALE, Lahan-Martins D, Almeida MFA, Lucchesi FR, Pacheco EO, Torres US, D'Ippolito G, Parente DB. Rare Malignant Liver Tumors: Current Insights and Imaging Challenges. Semin Ultrasound CT MR 2025:S0887-2171(25)00015-0. [PMID: 40220972 DOI: 10.1053/j.sult.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Rare malignant liver tumors (RMLTs) comprise a diverse group of neoplasms with distinct imaging features and significant diagnostic challenges due to their low prevalence and overlap with more common hepatic lesions. This review highlights the main radiologic characteristics of selected RMLTs-including fibrolamellar hepatocellular carcinoma, hepatic lymphoma, hepatocellular carcinoma in non-cirrhotic liver, mucinous cystic neoplasm, intraductal papillary neoplasm of the bile duct, epithelioid hemangioendothelioma, angiosarcoma, malignant hepatic adenoma, neuroendocrine tumor, hepatocholangiocarcinoma, hepatoblastoma, undifferentiated embryonal sarcoma, and infantile hepatic hemangioendothelioma-focusing on their presentation in CT and MRI. Recognizing specific imaging findings, such as arterial hyperenhancement, biliary communication, target and lollipop signs, and tumor morphology, can help narrow differential diagnoses and guide appropriate clinical management. Despite advancements in imaging, histopathologic confirmation is often required due to nonspecific features. Improved radiologic awareness of these rare entities is essential to facilitate early diagnosis and individualized treatment planning.
Collapse
Affiliation(s)
- Eleonora Salles-Silva
- Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; Grupo Fleury, Brazil
| | | | | | - Antonio Luis-Eiras de Araújo
- Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | | | | | | | | | - Ulysses S Torres
- Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil; Grupo Fleury, Brazil
| | - Giuseppe D'Ippolito
- Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil; Grupo Fleury, Brazil
| | - Daniella Braz Parente
- Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil; D'Or Institute for Research and Education, Rio de Janeiro, Brazil; Grupo Fleury, Brazil.
| |
Collapse
|
3
|
Zhang J, Tan Q, Fan Y, Xiao L, Zheng Z, Li K, Jing W, Song H, Liu X, Tan C, Wang X. Non-hypervascular pancreatic neuroendocrine neoplasms differentiation from CA19-9 negative pancreatic ductal adenocarcinomas based on contrast CT: A large sample series. Eur J Radiol 2025; 187:112095. [PMID: 40209484 DOI: 10.1016/j.ejrad.2025.112095] [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: 09/25/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
PROPOSE This study aims to evaluate the effectiveness of contrast-enhanced computed tomography (CT) in distinguishing non-hypervascular pancreatic neuroendocrine neoplasms (PNENs) from pancreatic ductal adenocarcinomas (PDACs) with a normal serum level of carbohydrate antigen 19-9 (CA19-9) levels. METHODS This retrospective study included 134 patients with pathologically confirmed non-hypervascular PNENs and 128 patients with CA19-9-negative PDACs, all of whom underwent contrast-enhanced CT prior to surgery between January 2015 and March 2024. Following independent evaluation by two radiologists, qualitative features from both groups were extracted in the arterial and portal venous phase and subsequently compared using univariate and multivariate analysis. RESULTS Patients with CA19-9 negative PDACs were significantly older than those with non-hypervascular PNENs (p < 0.001), and the majority of PDACs were located in the head of the pancreas (p < 0.01).Univariate analysis showed that non-hypervascular PNENs exhibited a higher frequency of well-defined tumor margins (p < 0.001) and calcification (p = 0.032) and a lower frequency of local invasion (p < 0.001), peripancreatic vascular invasion (p = 0.001), intra- or extrahepatic bile duct dilatation (p < 0.001), distal main pancreatic duct dilatation (p < 0.001), regional lymphadenopathy (p < 0.001) and tumor homogeneity (p < 0.001) when compared to CA19-9 negative PDACs. Multivariate analysis identified the absence of local invasion (Odds Ratio (OR) = 0.233; 95 % Confidence Internals (95 % CI):0.114-0.476; p < 0.001), absence of peripancreatic vascular invasion (OR = 0.434; 95 % CI:0.217-0.870; p = 0.019), a normal distal main pancreatic duct diameter (OR = 0.398; 95 % CI:0.202-0.785; p = 0.008), absence of regional lymphadenopathy (OR = 0.455; 95 % CI:0.238-0.870; p = 0.017) and tumor heterogeneity (OR = 0.240; 95 % CI:0.126-0.456; p < 0.001) as significant predictors of non-hypervascular PNENs. The area under the receiver operating characteristic curve for the radiological feature model was 0.829 based on logistic regression. CONCLUSIONS Qualitative features in contrast-enhanced CT images could be beneficial in differentially diagnosing non-hypervascular PNENs and CA19-9 negative PDACs.
Collapse
Affiliation(s)
- Jinyin Zhang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingquan Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Fan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Liu Xiao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenyi Jing
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haiyu Song
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan Province, China
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Xing Wang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
4
|
Lynch SE, Crawford CI, Houson HA, Omweri JM, Pukkanasut P, Gallegos CA, Whitt JD, Jaskula-Sztul R, Lapi SE, Sorace AG. Characterizing SSTR2 expression and modulation for targeted imaging and therapy in preclinical models of triple-negative breast cancer. Sci Rep 2025; 15:9988. [PMID: 40121305 PMCID: PMC11929780 DOI: 10.1038/s41598-025-94578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Patients with breast cancer which lack molecular targets, such as human epidermal growth factor receptor 2 (HER2) or hormone receptors, have limited access to targeted therapies. Somatostatin receptor 2 (SSTR2) is overexpressed in some cancers, and SSTR2-targeted radiopharmaceuticals are FDA-approved for theranostic targeted imaging and therapy in neuroendocrine tumors (NETs). Importantly, histone deacetylase (HDAC) inhibitors can epigenetically modulate SSTR2 expression in NETs with low or variable basal expression. The goal of this study is to characterize SSTR2 basal expression and induction via HDAC inhibition as a potential target for imaging and therapy in preclinical models of triple-negative breast cancer (TNBC). SSTR2 expression in mouse samples was assessed via Western blot and immunohistochemistry. Real-time quantitative PCR (qRT-PCR), flow cytometry, and cell binding assays were utilized to determine if HDAC inhibition can upregulate SSTR2 expression. [68Ga]Ga-DOTATATE positron emission tomography (PET) imaging, which targets SSTR2, was used to non-invasively characterize SSTR2 expression and variability in the EO771 and 4T1 TNBC models before and after HDAC inhibition. These studies demonstrate that HDAC inhibition can upregulate SSTR2 at the transcriptional, translational, and functional levels in breast cancer. Importantly, SSTR2 expression can be characterized non-invasively via PET imaging and modulation with HDAC inhibitors can be monitored longitudinally. Our findings highlight SSTR2 as a promising therapeutic molecular target in TNBC.
Collapse
Affiliation(s)
- Shannon E Lynch
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Corinne I Crawford
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey A Houson
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
| | - James M Omweri
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carlos A Gallegos
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason D Whitt
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Mamulashvili Bessac D, Baltzinger P, Poterszman N, Pham Van F, Collen C, Malouf GG, Ouvrard E, Kaseb A, Porot C, Ben Abdelghani M, Addeo P, Mertz L, Goichot B, Imperiale A. Organ-specific response to [ 177Lu]DOTATATE peptide receptor radionuclide therapy (PRRT) assessed by sequential [ 68Ga]DOTATOC PET/CT in patients with metastatic small intestine neuroendocrine tumors. Endocrine 2025; 87:1333-1341. [PMID: 39714577 DOI: 10.1007/s12020-024-04138-y] [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: 11/02/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
PURPOSE To evaluate organ-specific response to [177Lu]DOTATATE Peptide Receptor Radionuclide Therapy (PRRT) in patients with small intestine neuroendocrine tumor (SiNET) through [68Ga]DOTATOC PET/CT, and to analyze tumor uptake and functional volume variations at different metastatic sites in relation to disease progression during clinical follow-up after treatment. METHODS A retrospective analysis was conducted on 33 metastatic patients. PET/CT were performed pre-treatment (PET0), mid-treatment after two PRRT cycles (PET2), and post-treatment (PET4). SUVmax and somatostatin receptor-expressing tumor volume (SRETV) were measured in liver, lymph node, peritoneum/mesentery, and bone metastases. RESULTS Liver metastases showed significant reduction in both SUVmax and SRETV from PET0 to PET4, with early response evident after two PRRT cycles. Nodal lesions exhibited a delayed response, with significant reductions at PET4. Peritoneal and bone metastases showed a continuous decline in SUVmax, but no significant changes in SRETV. Objective response rates were highest in liver metastases after treatment completion with lesser responses in nodal, peritoneal, and bone lesions. At a median follow-up of 24.3 months, 70% of patients experienced disease progression, while 30% did not. Liver metastases showed no significant changes in SUVmax or SRETV regardless of progression. Conversely, in non-progressing patients, peritoneal/mesenteric lesions showed a significant reduction in SUVmax, with unchanged SRETV, and nodal metastases exhibited reduced SRETV. Bone lesions in non-progressing patients showed significant decreases in both SUVmax and SRETV. CONCLUSION [177Lu]DOTATATE PRRT effectively reduces tumor functional activity in patients with SiNETs, with organ-specific responses highlighting the need for personalized treatment strategies to optimize efficacy and minimize toxicity.
Collapse
Affiliation(s)
- Darejan Mamulashvili Bessac
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Philippe Baltzinger
- Endocrinology, Diabetology, Nutrition, University Hospitals of Strasbourg, Strasbourg University, Strasbourg, France
| | - Nathan Poterszman
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Floriane Pham Van
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Nuclear Medicine, Jean Minjoz University Hospital, Franche Compté University, Besançon, France
| | - Cedric Collen
- Nuclear Medicine, Jean Minjoz University Hospital, Franche Compté University, Besançon, France
| | - Gabriel G Malouf
- Oncology, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Ashjan Kaseb
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Radiology, College of Medicine, University of Jeddah, Jeddah, 23890, Saudi Arabia
| | - Clemence Porot
- Radiopharmacy, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Meher Ben Abdelghani
- Gastroenterology, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Pietro Addeo
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Hospitals of Strasbourg, Strasbourg, France
| | - Luc Mertz
- Radiophysics, University Hospitals of Strasbourg, Strasbourg, France
| | - Bernard Goichot
- Endocrinology, Diabetology, Nutrition, University Hospitals of Strasbourg, Strasbourg University, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France.
- IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France.
| |
Collapse
|
6
|
Kuiper J, Zoetelief E, Brabander T, de Herder WW, Hofland J. Current status of peptide receptor radionuclide therapy in grade 1 and 2 gastroenteropancreatic neuroendocrine tumours. J Neuroendocrinol 2025; 37:e13469. [PMID: 39563515 PMCID: PMC11919478 DOI: 10.1111/jne.13469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) using [177Lu-DOTA0,Tyr3]octreotate (177Lu-DOTATATE) represents an established treatment modality for somatostatin receptor-positive, locally advanced or metastatic gastroenteropancreatic neuroendocrine tumours (GEP NET) of grade 1 or 2. The studies have demonstrated that four cycles of PRRT with 177Lu-DOTATATE prolongs progression-free survival and preserves quality of life, in patients with grade 1 and 2 advanced GEP NET. Notably, first-line PRRT using 177Lu-DOTATATE in grade 2 and 3 GEP NET patients has also shown efficacy and safety. Furthermore, PRRT can ameliorate symptoms in patients with NET-associated functioning syndromes. Although various studies have explored alternative radionuclides for PRRT, none currently meet the criteria for routine clinical implementation. Ongoing research aims to further enhance PRRT, and the results from large clinical trials comparing PRRT with other NET treatments are anticipated, potentially leading to significant modifications in NET treatment strategies and PRRT protocols. The results of these studies are likely to help address existing knowledge gaps in the coming years. This review describes the clinical practice, recent developments and future treatment options of PRRT in patients with grade 1 and 2 GEP NET.
Collapse
Affiliation(s)
- Jelka Kuiper
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Eline Zoetelief
- Department of Radiology & Nuclear MedicineErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Tessa Brabander
- Department of Radiology & Nuclear MedicineErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Wouter W. de Herder
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Johannes Hofland
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| |
Collapse
|
7
|
Padwal MK, Parghane RV, Chakraborty A, Ujaoney AK, Anaganti N, Basu S, Basu B. Developing a peripheral blood RNA-seq based NETseq ensemble classifier: A potential novel tool for non-invasive detection and treatment response assessment in neuroendocrine tumor patients receiving 177Lu-DOTATATE PRRT. J Neuroendocrinol 2025; 37:e13462. [PMID: 39539072 PMCID: PMC11919474 DOI: 10.1111/jne.13462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/12/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Neuroendocrine tumors (NETs) are presented with metastases due to delayed diagnosis. We aimed to identify NET-related biomarkers from peripheral blood. The development and validation of a multi-gene NETseq ensemble classifier using peripheral blood RNA-Seq is reported. RNA-Seq was performed on peripheral blood samples from 178 NET patients and 73 healthy donors. Distinguishing gene features were identified from a learning cohort (59 PRRT-naïve GEP-NET patients and 38 healthy donors). Ensemble classifier combining the output of five machine learning algorithms viz. Random Forest (RF), Extreme Gradient Boosting (XGBOOST), Gradient Boosting Machine (GBM), Support Vector Machine (SVM), and Logistic Regression (LR) were trained and independently validated in the evaluation cohort (n = 106). The response to PRRT was evaluated in the PRRT cohort (n = 46) and the PRRT response monitoring cohort (n = 16). The response to 177Lu-DOTATATE PRRT was assessed using RECIST 1.1 criteria. The Ensemble classifier trained on 61 gene features, distinguished NET from healthy samples with 100% accuracy in the learning cohort. In an evaluation cohort, the classifier achieved 93% sensitivity (95% CI: 87.8%-98.03%) and 91.4% specificity (95% CI: 82.1%-100%) for PRRT-naïve GEP-NETs (AUROC = 95.4%). The classifier returned >87.5% sensitivity across different tumor characteristics and outperformed serum Chromogranin A sensitivity (χ2 = 21.89, p = 4.161e-6). In the PRRT cohort, RECIST 1.1 responders showed significantly lower NETseq prediction scores after 177Lu-DOTATATE PRRT, in comparison to the non-responders. In an independent response monitoring cohort, paired samples (before PRRT and after 2nd or 3rd cycle of PRRT) were analyzed. The NETseq prediction score significantly decreased in partial responders (p = .002) and marginally reduced in stable disease (p = .068). The NETseq ensemble classifier identified PRRT-naïve GEP-NETs with high accuracy (≥92%) and demonstrated a potential role in early treatment response monitoring in the PRRT setting. This blood-based, non-invasive, multi-analyte molecular method could be developed as a valuable adjunct to conventional methods in the detection and treatment response assessment in NET patients.
Collapse
Affiliation(s)
- Mahesh K. Padwal
- Molecular Biology DivisionBhabha Atomic Research CentreMumbaiIndia
- Homi Bhabha National InstituteMumbaiIndia
| | - Rahul V. Parghane
- Homi Bhabha National InstituteMumbaiIndia
- Radiation Medicine Centre, Bhabha Atomic Research CentreTata Memorial Centre AnnexeMumbaiIndia
| | - Avik Chakraborty
- Homi Bhabha National InstituteMumbaiIndia
- Radiation Medicine Centre, Bhabha Atomic Research CentreTata Memorial Centre AnnexeMumbaiIndia
| | | | - Narasimha Anaganti
- Molecular Biology DivisionBhabha Atomic Research CentreMumbaiIndia
- Homi Bhabha National InstituteMumbaiIndia
| | - Sandip Basu
- Homi Bhabha National InstituteMumbaiIndia
- Radiation Medicine Centre, Bhabha Atomic Research CentreTata Memorial Centre AnnexeMumbaiIndia
| | - Bhakti Basu
- Molecular Biology DivisionBhabha Atomic Research CentreMumbaiIndia
- Homi Bhabha National InstituteMumbaiIndia
| |
Collapse
|
8
|
Zhao J, Pei X, Li Y. Efficacy and Safety of Peptide Receptor Radionuclide Therapy for the Treatment of Pancreatic Neuroendocrine Tumors: A Systematic Review and Meta-Analysis of Comparative Studies. Cureus 2025; 17:e80817. [PMID: 40190903 PMCID: PMC11970540 DOI: 10.7759/cureus.80817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) showed promising potential in the management of patients with pancreatic neuroendocrine tumors (pNETs). However, there is still a lack of evidence on its relative efficacy and safety compared with other treatment options. This review aims to synthesize the existing evidence on the efficacy and safety of PRRT for pNETs compared to different treatments. An electronic search was conducted from inception to May 2024. Comparative studies, including randomized controlled trials (RCTs), cohort studies, and case-control studies, that focused on the use of PRRT for treating pNETs were included. Efficacy outcomes included disease control rate (DCR), complete response (CR), partial response (PR), stable disease (SD), progression-free survival (PFS), and overall survival (OS). Safety outcomes were grade 3-4 hematological and renal toxicity and adverse events (AEs). Nine studies met the inclusive criteria. Among them, only one (11.1%) study was an RCT. Meta-analysis between full and reduced dosages of 177Lu-DOTATATE for G1-G2 pNETs revealed no significant differences in DCR, CR, PR, SD, and PFS between the groups. However, the full dosage group showed superior efficacy in some outcomes (DCR, CR, and PR). When PRRT was compared to other treatments such as surgery, chemotherapy, and targeted agents, it was associated with longer PFS and OS. Additionally, PRRT combined with capecitabine and salvage PRRT also showed efficacy in advanced cases. Safety analysis indicated that PRRT is well-tolerated, with minimal severe toxicity reported. PRRT is a promising therapeutic option for patients with advanced pNETs, offering a balance of efficacy and safety compared to other available treatments based on the low quality of evidence. Full-dosage PRRT may provide better outcomes than reduced dosages, and salvage PRRT remains effective for progressive disease. However, further high-quality RCTs are needed to confirm these findings and optimize PRRT usage in pNETs.
Collapse
Affiliation(s)
- Jun Zhao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, CHN
| | - Xiaxia Pei
- Department of Oncology, Second Hospital of Lanzhou University, Lanzhou, CHN
| | - Yumin Li
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, CHN
| |
Collapse
|
9
|
Satapathy S, Aggarwal P, Sood A, Chandekar KR, Das CK, Gupta R, Khosla D, Das N, Kapoor R, Kumar R, Singh H, Shukla J, Kumar A, Mittal BR. 177Lu-DOTATATE Plus Capecitabine Versus 177Lu-DOTATATE Alone in Patients with Advanced Grade 1/2 Gastroenteropancreatic Neuroendocrine Tumors (LuCAP): A Randomized, Phase 2 Trial. J Nucl Med 2025; 66:238-244. [PMID: 39778968 DOI: 10.2967/jnumed.124.268617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
177Lu-DOTATATE has emerged as a viable treatment strategy for advanced well-differentiated grade 1/2 gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Few retrospective studies have shown concomitant 177Lu-DOTATATE with radiosensitizing low-dose capecitabine to be effective in advanced NETs. However, this has not been validated in prospective randomized-controlled trials. Methods: In this investigator-initiated, parallel-group, open-label, phase 2 trial, patients with grade 1/2 GEP-NETs, having progressive somatostatin receptor-positive, locally advanced, or metastatic disease on 68Ga-DOTANOC PET/CT, were randomly assigned in a 1:1 ratio to 177Lu-DOTATATE plus capecitabine (experimental arm) or 177Lu-DOTATATE only (control arm). 177Lu-DOTATATE was administered at approximately 7.4 GBq/cycle intravenously, for up to 4 cycles, at 8 wk intervals, whereas capecitabine was given at 1,250 mg/m2/d orally from day 0 to day 14 of each cycle of 177Lu-DOTATATE. The primary endpoint was the objective response rate. Secondary endpoints included the disease control rate, progression-free survival, overall survival, and adverse events. Results: Seventy-two patients (median age, 53 y; range, 18-79 y) were enrolled. The objective response rate was 33.3% (95% CI, 18.6-50.9%) in the experimental arm versus 30.6% (95% CI, 16.4-48.1%) in the control arm (P = 0.800). The disease control rate was 88.9% (95% CI, 73.9-96.9%) and 91.7% (95% CI, 77.5-98.2%) in the experimental and control arms, respectively (P = 1.000). The estimated median progression-free survival in the experimental arm was 29 mo (95% CI, 22-29 mo) versus 31 mo (95% CI, 29-32 mo) in the control arm (P = 0.401). The median overall survival was not reached in either arm (P = 0.876). Overall, adverse events of at least grade 3 were noted in 7 patients in the experimental arm versus 6 patients in the control arm (P = 0.759). Conclusion: Based on the results of this trial, the addition of low-dose capecitabine to 177Lu-DOTATATE in advanced grade 1/2 GEP-NETs did not lead to superior radiographic responses. Further studies are needed to evaluate its potential role in high-grade NETs.
Collapse
Affiliation(s)
- Swayamjeet Satapathy
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Piyush Aggarwal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashwani Sood
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India;
| | - Kunal R Chandekar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chandan K Das
- Department of Clinical Haematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
- Montefiore Medical Center, Bronx, New York
| | - Rajesh Gupta
- Department of GI Surgery, HPB and Liver Transplantation, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Divya Khosla
- Department of Radiotherapy and Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India; and
| | - Namrata Das
- Department of Radiotherapy and Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India; and
- Proton International London Ltd., University College London Hospitals, London, United Kingdom
| | - Rakesh Kapoor
- Department of Radiotherapy and Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India; and
| | - Rajender Kumar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harmandeep Singh
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Kumar
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
10
|
Yao Z, Chen H. Everolimus in pituitary tumor: a review of preclinical and clinical evidence. Front Endocrinol (Lausanne) 2024; 15:1456922. [PMID: 39736867 PMCID: PMC11682973 DOI: 10.3389/fendo.2024.1456922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Although pituitary tumors (PTs) are mostly benign, some PTs are characterized by low surgical resection rates, high recurrence rates, and poor response to conventional treatments and profoundly affect patients' quality of life. Everolimus (EVE) is the only FDA-approved mTOR inhibitor, which can be used for oral treatment. It effectively inhibits tumor cell proliferation and angiogenesis. It has been administered for various neuroendocrine tumors of the digestive tract, lungs, and pancreas. EVE not only suppresses the growth and proliferation of APT cells but also enhances their sensitivity to radiotherapy and chemotherapy. This review introduces the role of the PI3K/AKT/mTOR pathway in the development of APTs, comprehensively explores the current status of preclinical and clinical research of EVE in APTs, and discusses the blood-brain barrier permeability and safety of EVE.
Collapse
Affiliation(s)
- Zihong Yao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hui Chen
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Xiao N, Zhu S, Liu H, Li L, Shi L. Treatment and management of duodenal gangliocytic paraganglioma: A case report. Exp Ther Med 2024; 28:435. [PMID: 39355522 PMCID: PMC11443592 DOI: 10.3892/etm.2024.12723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/15/2024] [Indexed: 10/03/2024] Open
Abstract
Gangliocytic paraganglioma (GP) is a rare neuroendocrine tumor primarily found in the duodenum, most commonly in the second and third sections of the duodenum. Diagnosis of GP is based on its distinctive histopathological characteristics, which include three types of tumor cells in varying proportions: i) Epithelioid, ii) spindle-like and iii) ganglion-shaped cells. The distribution of the three tumor cell components varies from case to case and a patient may be easily misdiagnosed if one of the components is predominant. Endoscopic submucosal dissection (ESD) or surgical resection is the ideal treatment for duodenal GP (DGP); however, biotherapy, nuclide therapy, chemotherapy, targeted therapy and immunotherapy can be selected individually for patients with postoperative recurrence, metastasis or not suitable for surgery. In the present study, a male patient with DGP experienced recurrence after ESD surgery, and so received octreotide (Novartis; 30 mg/28 days) for 12 consecutive cycles. The patient had no further symptoms of gastrointestinal bleeding and no new lesions or metastases were observed after 47 months of follow-up.
Collapse
Affiliation(s)
- Na Xiao
- Department of Gastroenterology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Sumin Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Huan Liu
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou, Jiangsu, 221002, P.R. China
| | - Longfei Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Lihong Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
12
|
Fields BC, Ayabe RI, Seo YD, Maxwell JE, Halperin DM. Current Status of Immunotherapy in Management of Small Bowel Neuroendocrine Tumors. Curr Oncol Rep 2024; 26:1530-1542. [PMID: 39466478 PMCID: PMC11776107 DOI: 10.1007/s11912-024-01610-w] [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: 09/30/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW This study aims to present the current landscape of immunotherapy in the management of small bowel neuroendocrine tumors and identify ongoing and future targets for improved response. RECENT FINDINGS Somatostatin analogs and mTOR inhibitors remain cornerstones of non-surgical treatment, and applications of PRRT in SBNET are promising. Several efforts to replicate the success of immunotherapies in other solid tumors have been attempted in SBNET, with limited responses observed with current immune targets, such as PD-1/PD-L1 and CTLA-4. Epigenetic analyses have suggested a potential role for methylation and histone acetylation in SBNET tumorigenesis that warrant greater exploration. While the incidence of SBNET continues to increase, the number of effective therapies is few. Further elucidation of targetable components of the SBNET immune microenvironment with greater modulatory effects is necessary to improve outcomes in this growing patient population.
Collapse
Affiliation(s)
- Brittany C Fields
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Reed I Ayabe
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Y David Seo
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jessica E Maxwell
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Daniel M Halperin
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
13
|
Perry RR, Feliberti EC, Hughes MS. Management of Pancreatic Neuroendocrine Tumors: Surgical Strategies and Controversies. Endocr Pract 2024; 30:908-916. [PMID: 39032831 DOI: 10.1016/j.eprac.2024.07.010] [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: 03/05/2024] [Revised: 06/01/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE Pancreatic neuroendocrine tumors (PNETs) are uncommon tumors which are increasing in incidence. The management of these tumors continues to evolve. This review examines the current role of surgery in the treatment of these tumors. METHODS Studies published over the past 10 years were identified using several databases including PubMed, MEDLINE, and Science Direct. Search terms included PNETs, treatment, and surgery. Clinical practice guidelines and updates from several major groups were reviewed. RESULTS Surgery continues to have a major role in the treatment of sporadic functional and nonfunctional PNETs. Pancreas-sparing approaches are increasingly accepted as alternatives to formal pancreatic resection in selected patients. Options such as watch and wait or endoscopic ablation may be reasonable alternatives to surgery for non-functional PNETs < 2 cm in size. Surgical decision-making in multiple endocrine neoplasia type 1 patients remains complex and in some situations such as gastrinoma quite controversial. The role of surgery has significantly diminished in patients with advanced disease due to the advent of more effective systemic and liver-directed therapies. However, the optimal treatments and sequencing in advanced disease remain poorly defined, and it has been suggested that surgery is underutilized in these patients. CONCLUSIONS Surgery remains a major treatment modality for PNETs. Given the plethora of available treatments, ongoing controversies and the changing landscape, management has become increasingly complex. An experienced multidisciplinary team which includes surgery is essential to manage these patients.
Collapse
Affiliation(s)
- Roger R Perry
- Division of Surgical Oncology, Eastern Virginia Medical School, Norfolk, Virginia.
| | - Eric C Feliberti
- Division of Surgical Oncology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Marybeth S Hughes
- Division of Surgical Oncology, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
14
|
Tan B, Zhang B, Chen H. Gastroenteropancreatic neuroendocrine neoplasms: epidemiology, genetics, and treatment. Front Endocrinol (Lausanne) 2024; 15:1424839. [PMID: 39411312 PMCID: PMC11474919 DOI: 10.3389/fendo.2024.1424839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
The incidence of gastroenteropancreatic neuroendocrine neoplasms (GEP NEN) is increasing at a rapid pace and is becoming an increasingly important consideration in clinical care. Epidemiological data from multiple countries indicate that the incidence of gastroenteropancreatic neuroendocrine neoplasms (GEP NEN) exhibits regional, site-specific, and gender-based variations. While the genetics and pathogenesis of some GEP NEN, particularly pancreatic NENs, have been investigated, there are still many mechanisms that require further investigation. The management of GEP NEN is diverse, but surgery remains the primary option for most cases. Peptide receptor radionuclide therapy (PRRT) is an effective treatment, and several clinical trials are exploring the potential of immunotherapy and targeted therapy, as well as combination therapy.
Collapse
Affiliation(s)
- Baizhou Tan
- Department of Histology and Embryology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Beiyu Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongping Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory of Experimental Animals, Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Grewal US, Loeffler BT, Paschke A, Dillon JS, Chandrasekharan C. Repeat Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumors: A NET Center of Excellence Experience. J Gastrointest Cancer 2024; 55:1165-1170. [PMID: 38780680 DOI: 10.1007/s12029-024-01065-z] [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: 05/05/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION The available data for the safety and efficacy of repeat peptide receptor radionuclide therapy (PRRT) are almost exclusively from European centers. We present an updated experience with repeat PRRT in a cohort of US patients with neuroendocrine tumors (NETs) at our NET center of excellence. METHODS We used our single-center longitudinal NET registry to identify patients who had been previously treated with at least one dose of PRRT (PRRT 1, either 177Lu DOTATATE or 90Y DOTATOC) and following radiographic disease progression were re-treated with a second course of PRRT (PRRT 2). We reviewed patient, tumor and treatment characteristics, objective response rates, and toxicities after PRRT 1 and PRRT 2. RESULTS A total of 11 patients were included in the analysis. 45.5% (5/11) of patients received 177Lu DOTATATE PRRT only, both for PRRT1 and PRRT 2, while 54.5% (6/11) of patients received 90Y DOTATOC PRRT for PRRT1. At first restaging scan after PRRT2 (3-6 months), 18.2% (2/11), 36.4% (4/11), and 27.3% (3/11) of patients had PR, SD, and PD, respectively; 2/11 patients (18.2%) died before the first restaging scan. Therefore, 5/11 (45.5%) patients were noted to have disease progression. Median PFS for PRRT1 was 25.4 months and median PFS for PRRT2 was 13.1 months (p = 0.0001). We did not find a statistically significant difference between the occurrence of short and long-term hematological toxicities as well as renal toxicity after PRRT1 and PRRT2. CONCLUSION We show that repeat PRRT may benefit select patients and have an acceptable safety profile. In our cohort, PFS was significantly lower after PRRT2 as compared to PRRT1.
Collapse
Affiliation(s)
- Udhayvir S Grewal
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Bradley T Loeffler
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Alexander Paschke
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Joseph S Dillon
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, IA, USA
- Division of Endocrinology and Metabolism, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, ENETS Center of Excellence, Iowa City, IA, USA
| | - Chandrikha Chandrasekharan
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, ENETS Center of Excellence, Iowa City, IA, USA.
| |
Collapse
|
16
|
Panzuto F, Lamarca A, Fazio N. Comparative analysis of international guidelines on the management of advanced non-functioning well-differentiated pancreatic neuroendocrine tumors. Cancer Treat Rev 2024; 129:102803. [PMID: 39029154 DOI: 10.1016/j.ctrv.2024.102803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
This review presents a comprehensive comparative analysis of international guidelines for managing advanced, non-functioning, well-differentiated pancreatic neuroendocrine tumors (panNETs). PanNETs, which represent a significant proportion of pancreatic neuroendocrine neoplasms, exhibit diverse clinical behaviors and prognoses based on differentiation, grading, and other molecular markers. The varying therapeutic strategies proposed by different guidelines reflect their distinct emphases and regional considerations, such as the ESMO guideline's focus on advanced disease management and the ENETS guidance paper's multidisciplinary approach. This review examines the most recent guidelines from ESMO, NCCN, ASCO, ENETS, and NANETS, analyzing the recommendations for first-line therapies and subsequent treatment pathways in different clinical scenarios. Significant variations are observed in the recommendations, particularly concerning the choice and sequence of systemic therapies, the role of tumor grading and the Ki-67 index in therapeutic decisions, and the integration of regional regulatory and clinical practices. The analysis highlights the need for a tailored approach to managing advanced NF panNETs, advocating for flexibility in applying guidelines to account for individual patient circumstances and the evolving evidence base. This work underscores the complexities of managing this patient population and the critical role of a multidisciplinary team in optimizing treatment outcomes.
Collapse
Affiliation(s)
- Francesco Panzuto
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Digestive Disease Unit, Sant'Andrea University Hospital ENETS Center of Excellence, Rome, Italy.
| | - Angela Lamarca
- Department of Medical Oncology, Oncohealth Institute, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, IT, Italy
| |
Collapse
|
17
|
Ghabra S, Ramamoorthy B, Andrews SG, Sadowski SM. Surgical Management and Long-Term Evaluation of Pancreatic Neuroendocrine Tumors. Surg Clin North Am 2024; 104:891-908. [PMID: 38944507 PMCID: PMC11214659 DOI: 10.1016/j.suc.2024.02.019] [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] [Indexed: 07/01/2024]
Abstract
Pancreatic neuroendocrine tumors (PNETs) arise from neuroendocrine cells and are a rare class of heterogenous tumors with increasing incidence. The diagnosis, staging, treatment, and prognosis of PNETs depend heavily on identifying the histologic features and biological mechanisms. Here, the authors provide an overview of the diagnostic workup (biomarkers and imaging), grade, and staging of PNETs. The authors also explore associated genetic mutations and molecular pathways and describe updated guidelines on surgical and systemic treatment modalities.
Collapse
Affiliation(s)
- Shadin Ghabra
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. https://twitter.com/ShadinGhabra_MD
| | - Bhavishya Ramamoorthy
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen G Andrews
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 CRC, Room 4-5932, Bethesda, MD 20892, USA. https://twitter.com/AndrewsStephenG
| | - Samira M Sadowski
- Neuroendocrine Cancer Therapy Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 CRC, Room 4-5932, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Chauhan A, Chan K, Halfdanarson TR, Bellizzi AM, Rindi G, O’Toole D, Ge PS, Jain D, Dasari A, Anaya DA, Bergsland E, Mittra E, Wei AC, Hope TA, Kendi AT, Thomas SM, Flem S, Brierley J, Asare EA, Washington K, Shi C. Critical updates in neuroendocrine tumors: Version 9 American Joint Committee on Cancer staging system for gastroenteropancreatic neuroendocrine tumors. CA Cancer J Clin 2024; 74:359-367. [PMID: 38685134 PMCID: PMC11938941 DOI: 10.3322/caac.21840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 05/02/2024] Open
Abstract
The American Joint Committee on Cancer (AJCC) staging system for all cancer sites, including gastroenteropancreatic neuroendocrine tumors (GEP-NETs), is meant to be dynamic, requiring periodic updates to optimize AJCC staging definitions. This entails the collaboration of experts charged with evaluating new evidence that supports changes to each staging system. GEP-NETs are the second most prevalent neoplasm of gastrointestinal origin after colorectal cancer. Since publication of the AJCC eighth edition, the World Health Organization has updated the classification and separates grade 3 GEP-NETs from poorly differentiated neuroendocrine carcinoma. In addition, because of major advancements in diagnostic and therapeutic technologies for GEP-NETs, AJCC version 9 advocates against the use of serum chromogranin A for the diagnosis and monitoring of GEP-NETs. Furthermore, AJCC version 9 recognizes the increasing role of endoscopy and endoscopic resection in the diagnosis and management of NETs, particularly in the stomach, duodenum, and colorectum. Finally, T1NXM0 has been added to stage I in these disease sites as well as in the appendix.
Collapse
Affiliation(s)
- Aman Chauhan
- Department of Medicine, Neuroendocrine Oncology, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | - Kelley Chan
- Department of Surgery, Loyola University Medical Center, Chicago, Illinois, USA
| | | | - Andrew M. Bellizzi
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA, USA
| | - Guido Rindi
- Department of Life Sciences, Section of Anatomic Pathology, Università Cattolica del Sacro Cuore; Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS and Roma-Gemelli ENETS Center of Excellence, Roma, Italy
| | - Dermot O’Toole
- National Centre for Neuroendocrine Tumours, ENETS Centre of Excellence (St. Vincent’s University Hospital) and St. James Hospital, Trinity College Dublin, Dublin, Ireland
| | - Phillip S. Ge
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dhanpat Jain
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel A. Anaya
- Department of Gastrointestinal Oncology-Surgery, Moffitt Cancer Center, Tampa, FL, USA
| | - Emily Bergsland
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Erik Mittra
- Department of Diagnostic Radiology, Molecular Imaging and Therapy, Oregon Health &Science University, Portland, Oregon, USA
| | - Alice C. Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Thomas A. Hope
- Department of Radiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ayse T. Kendi
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Samantha M. Thomas
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Sherlonda Flem
- Tumor Registrar, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - James Brierley
- Radiation Medicine Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Elliot A. Asare
- Department of Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Chanjuan Shi
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
19
|
Violante T, Murphy B, Ferrari D, Graham RP, Navin P, Merchea A, Larson DW, Dozois EJ, Halfdanarson TR, Perry WR. Presacral Neuroendocrine Neoplasms: A Multi-site Review of Surgical Outcomes. Ann Surg Oncol 2024; 31:4551-4557. [PMID: 38679679 DOI: 10.1245/s10434-024-15328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/01/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Presacral neuroendocrine neoplasms (PNENs) are rare tumors, with limited data on management and outcomes. METHODS A retrospective review of institutional medical records was conducted to identify all patients with PNENs between 2008 and 2022. Data collection included demographics, symptoms, imaging, surgical approaches, pathology, complications, and long-term outcomes. RESULTS Twelve patients were identified; two-thirds were female, averaging 44.8 years of age, and, for the most part, presenting with back pain, constipation, and abdominal discomfort. Preoperative imaging included computed tomography scans and magnetic resonance images, with somatostatin receptor imaging and biopsies being common. Half of the patients had metastatic disease on presentation. Surgical approach varied, with anterior, posterior, and combined techniques used, often involving muscle transection and coccygectomy. Short-term complications affected one-quarter of patients. Pathologically, PNENs were mainly well-differentiated grade 2 tumors with positive synaptophysin and chromogranin A. Associated anomalies were common, with tail-gut cysts prevalent. Mean tumor diameter was 6.3 cm. Four patients received long-term adjuvant therapy. Disease progression necessitated additional interventions, including surgery and various chemotherapy regimens. Skeletal, liver, thyroid, lung, and pancreatic metastases occurred during follow-up, with no mortality reported. Kaplan-Meier analysis showed a 5-year local recurrence rate of 23.8%, disease progression rate of 14.3%, and de novo metastases rate of 30%. CONCLUSION The study underscores the complex management of PNENs and emphasizes the need for multicenter research to better understand and manage these tumors. It provides valuable insights into surgical outcomes, recurrence rates, and overall survival, guiding future treatment strategies for PNEN patients.
Collapse
Affiliation(s)
- Tommaso Violante
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
- School of General Surgery, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Brenda Murphy
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - Davide Ferrari
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
- School of General Surgery, Alma Mater Studiorum University of Bologna, Bologna, Italy
- General Surgery Residency Program, University of Milan, Milan, Italy
| | - Rondell P Graham
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Patrick Navin
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Amit Merchea
- Division of Colon and Rectal Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - David W Larson
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | - Eric J Dozois
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - William R Perry
- Division of Colon and Rectal Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Singh S, Halperin D, Myrehaug S, Herrmann K, Pavel M, Kunz PL, Chasen B, Tafuto S, Lastoria S, Capdevila J, García-Burillo A, Oh DY, Yoo C, Halfdanarson TR, Falk S, Folitar I, Zhang Y, Aimone P, de Herder WW, Ferone D. [ 177Lu]Lu-DOTA-TATE plus long-acting octreotide versus high‑dose long-acting octreotide for the treatment of newly diagnosed, advanced grade 2-3, well-differentiated, gastroenteropancreatic neuroendocrine tumours (NETTER-2): an open-label, randomised, phase 3 study. Lancet 2024; 403:2807-2817. [PMID: 38851203 DOI: 10.1016/s0140-6736(24)00701-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND There are currently no standard first-line treatment options for patients with higher grade 2-3, well-differentiated, advanced, gastroenteropancreatic neuroendocrine tumours. We aimed to investigate the efficacy and safety of first-line [177Lu]Lu-DOTA-TATE (177Lu-Dotatate) treatment. METHODS NETTER-2 was an open-label, randomised, parallel-group, superiority, phase 3 trial. We enrolled patients (aged ≥15 years) with newly diagnosed higher grade 2 (Ki67 ≥10% and ≤20%) and grade 3 (Ki67 >20% and ≤55%), somatostatin receptor-positive (in all target lesions), advanced gastroenteropancreatic neuroendocrine tumours from 45 centres across nine countries in North America, Europe, and Asia. We used interactive response technologies to randomly assign (2:1) patients to receive four cycles (cycle interval was 8 weeks ± 1 week) of intravenous 177Lu-Dotatate plus intramuscular octreotide 30 mg long-acting repeatable (LAR) then octreotide 30 mg LAR every 4 weeks (177Lu-Dotatate group) or high-dose octreotide 60 mg LAR every 4 weeks (control group), stratified by neuroendocrine tumour grade (2 vs 3) and origin (pancreas vs other). Tumour assessments were done at baseline, week 16, and week 24, and then every 12 weeks until disease progression or death. The primary endpoint was progression-free survival by blinded, independent, central radiology assessment. We did the primary analysis at 101 progression-free survival events as the final progression-free survival analysis. NETTER-2 is registered with ClinicalTrials.gov, NCT03972488, and is active and not recruiting. FINDINGS Between Jan 22, 2020, and Oct 13, 2022, we screened 261 patients, 35 (13%) of whom were excluded. We randomly assigned 226 (87%) patients (121 [54%] male and 105 [46%] female) to the 177Lu-Dotatate group (n=151 [67%]) and control group (n=75 [33%]). Median progression-free survival was 8·5 months (95% CI 7·7-13·8) in the control group and 22·8 months (19·4-not estimated) in the 177Lu-Dotatate group (stratified hazard ratio 0·276 [0·182-0·418]; p<0·0001). During the treatment period, adverse events (of any grade) occurred in 136 (93%) of 147 treated patients in the 177Lu-Dotatate group and 69 (95%) of 73 treated patients in the control group. There were no study drug-related deaths during the treatment period. INTERPRETATION First-line 177Lu-Dotatate plus octreotide LAR significantly extended median progression-free survival (by 14 months) in patients with grade 2 or 3 advanced gastroenteropancreatic neuroendocrine tumours. 177Lu-Dotatate should be considered a new standard of care in first-line therapy in this population. FUNDING Advanced Accelerator Applications, a Novartis Company.
Collapse
Affiliation(s)
- Simron Singh
- University of Toronto, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada.
| | | | - Sten Myrehaug
- University of Toronto, Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; National Center for Tumor Diseases (NCT), NCT West, Heidelberg, Germany
| | - Marianne Pavel
- Department of Medicine 1, Uniklinikum Erlangen, and Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Pamela L Kunz
- Yale School of Medicine and Yale Cancer Center, Yale University, New Haven, CT, USA
| | | | - Salvatore Tafuto
- Sarcoma and Rare Tumors Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Secondo Lastoria
- Division of Nuclear Medicine, Istituto Nazionale Tumori IRCCS, Fondazione G Pascale, Naples, Italy
| | - Jaume Capdevila
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Amparo García-Burillo
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Changhoon Yoo
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Stephen Falk
- Bristol Haematology and Oncology Centre, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | | | - Yufen Zhang
- Novartis Pharmaceuticals Corp, East Hanover, NJ, USA
| | | | | | - Diego Ferone
- Endocrinology, IRCCS Policlinico San Martino and DiMI, University of Genova, Genoa, Italy
| |
Collapse
|
21
|
Hoogenkamp DS, de Wit-van der Veen LJ, Huizing DMV, Tesselaar MET, van Leeuwaarde RS, Stokkel MPM, Lam MGEH, Braat AJAT. Advances in Radionuclide Therapies for Patients with Neuro-endocrine Tumors. Curr Oncol Rep 2024; 26:551-561. [PMID: 38598035 PMCID: PMC11062977 DOI: 10.1007/s11912-024-01521-w] [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] [Accepted: 03/21/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE OF REVIEW To provide insights into the role of peptide receptor radionuclide therapy (PRRT) in patients with advanced neuroendocrine tumors (NET) and an overview of possible strategies to combine PRRT with locoregional and systemic anticancer treatments. RECENT FINDINGS Research on combining PRRT with other treatments encompasses a wide variety or treatments, both local (transarterial radioembolization) and systemic therapies, chemotherapy (i.e., capecitabine and temozolomide), targeted therapies (i.e., olaparib, everolimus, and sunitinib), and immunotherapies (e.g., nivolumab and pembrolizumab). Furthermore, PRRT shows promising first results as a treatment prior to surgery. There is great demand to enhance the efficacy of PRRT through combination with other anticancer treatments. While research in this area is currently limited, the field is rapidly evolving with numerous ongoing clinical trials aiming to address this need and explore novel therapeutic combinations.
Collapse
Affiliation(s)
- Denise S Hoogenkamp
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Linda J de Wit-van der Veen
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Daphne M V Huizing
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Margot E T Tesselaar
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachel S van Leeuwaarde
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Marnix G E H Lam
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
22
|
Carsote M, Nistor C. Neuroendocrine Neoplasia: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2024; 12:801. [PMID: 38672156 PMCID: PMC11048153 DOI: 10.3390/biomedicines12040801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Neuroendocrine neoplasia (NEN) represents a sensational field of modern medicine; immense progress in emerging biochemical, molecular, endocrine, immunohistochemical, and serum tumour markers of disease, respectively, which are part of early diagnosis, genetic testing, and multidisciplinary approaches [...].
Collapse
Affiliation(s)
- Mara Carsote
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Clinical Endocrinology V, “C.I. Parhon” National Institute of Endocrinology, 011863 Bucharest, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Thoracic Surgery Department, “Dr. Carol Davila” Central Military University Emergency Hospital, 010242 Bucharest, Romania
| |
Collapse
|
23
|
Arrivi G, Specchia M, Pilozzi E, Rinzivillo M, Caruso D, Santangeli C, Prosperi D, Ascolese AM, Panzuto F, Mazzuca F. Diagnostic and Therapeutic Management of Primary Orbital Neuroendocrine Tumors (NETs): Systematic Literature Review and Clinical Case Presentation. Biomedicines 2024; 12:379. [PMID: 38397981 PMCID: PMC10886459 DOI: 10.3390/biomedicines12020379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The ocular involvement of neuroendocrine neoplasms (NENs) is uncommon and mainly represented by metastases from gastrointestinal and lung neuroendocrine tumors. Primary orbital NENs are even less common and their diagnostic and therapeutic management is a challenge. METHODS A systematic review of the literature was conducted from 1966 to September 2023 on PubMed to identify articles on orbital NENs and to summarize their clinical-pathological features, diagnosis and therapeutic management. Furthermore, we presented a case of a locally advanced retro-orbital primary neuroendocrine tumor that was referred to the certified Center of Excellence of Sant'Andrea Hospital, La Sapienza University of Rome, Italy. RESULTS The final analysis included 63 records on orbital NENs and 11 records focused on primary orbital NENs. The localization was mostly unilateral and in the right orbit; proptosis or exophthalmos represented the initial symptoms. The diagnostic work-up and therapeutic management was discussed and a diagnostic algorithm for the suspicion of primary orbital NENs was proposed. CONCLUSIONS A multidisciplinary approach is required for the management of primary orbital NENs, emphasizing the importance of early referral to dedicated centers for prompt differential diagnosis, tailored treatment, and an improved quality of life and survival.
Collapse
Affiliation(s)
- Giulia Arrivi
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Grottarossa Street 1035-1039, 00189 Rome, Italy; (M.S.); (F.M.)
- Department of Medical and Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, PhD School in Translational Medicine and Oncology, Sapienza University of Rome, 00189 Rome, Italy
| | - Monia Specchia
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Grottarossa Street 1035-1039, 00189 Rome, Italy; (M.S.); (F.M.)
| | - Emanuela Pilozzi
- Anatomia Patologica Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| | - Maria Rinzivillo
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Digestive Disease Unit, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
| | - Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (D.C.); (C.S.)
| | - Curzio Santangeli
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (D.C.); (C.S.)
| | - Daniela Prosperi
- Nuclear Medicine Unit, Sant’Andrea University Hospital, 00189 Rome, Italy;
| | - Anna Maria Ascolese
- Radiotherapy Oncology Unit, Department of Surgical Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy;
| | - Francesco Panzuto
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Digestive Disease Unit, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
- European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Federica Mazzuca
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Grottarossa Street 1035-1039, 00189 Rome, Italy; (M.S.); (F.M.)
| |
Collapse
|