1
|
Tacelli M, Gentiluomo M, Biamonte P, Castano JP, Berković MC, Cives M, Kapitanović S, Marinoni I, Marinovic S, Nikas I, Nosáková L, Pedraza-Arevalo S, Pellè E, Perren A, Strosberg J, Campa D, Capurso G. Pancreatic neuroendocrine neoplasms (pNENs): Genetic and environmental biomarkers for risk of occurrence and prognosis. Semin Cancer Biol 2025; 112:112-125. [PMID: 40158764 DOI: 10.1016/j.semcancer.2025.03.005] [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: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are rare and heterogeneous tumors arising from neuroendocrine cells, representing approximately 10 % of all Gastro-Entero-Pancreatic neuroendocrine neoplasms. While most pNENs are sporadic, a subset is associated with genetic syndromes such as multiple endocrine neoplasia type 1 (MEN1) or von Hippel-Lindau disease (VHL). pNENs are further classified into functioning and non-functioning tumors, with distinct clinical behaviors, prognoses, and treatment approaches. This review explores genetic and environmental biomarkers that influence the risk, prognosis, and therapeutic responses in pNENs. The epidemiology of pNENs reveals an increasing incidence, primarily due to advancements in imaging techniques. Genetic factors play a pivotal role, with germline mutations in MEN1, VHL, and other genes contributing to familial pNENs. Somatic mutations, including alterations in the mTOR pathway and DNA maintenance genes such as DAXX and ATRX, are critical in sporadic pNENs. These mutations, along with epigenetic dysregulation and transcriptomic alterations, underpin the diverse clinical and molecular phenotypes of pNENs. Emerging evidence suggests that epigenetic changes, including DNA methylation profiles, can stratify pNEN subtypes and predict disease progression. Environmental and lifestyle factors, such as diabetes, smoking, and chronic pancreatitis, have been linked to an increased risk of sporadic pNENs. While the association between these factors and tumor progression is still under investigation, their potential role in influencing therapeutic outcomes warrants further study. Advances in systemic therapies, including somatostatin analogs, mTOR inhibitors, and tyrosine kinase inhibitors, have improved disease management. Biomarkers such as Ki-67, somatostatin receptor expression, and O6-methylguanine-DNA methyltransferase (MGMT) status are being evaluated for their predictive value. Novel approaches, including the use of circulating biomarkers (NETest, circulating tumor cells, and ctDNA) and polygenic risk scores, offer promising avenues for non-invasive diagnosis and monitoring. Despite these advancements, challenges remain, including the need for large, well-annotated datasets and validated biomarkers. Future research should integrate multi-omics approaches and leverage liquid biopsy technologies to refine diagnostic, prognostic, and therapeutic strategies. Interdisciplinary collaborations and global consortia are crucial for overcoming current limitations and translating research findings into clinical practice. These insights hold promise for improving prevention, early detection, and tailored treatments, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Paolo Biamonte
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Justo P Castano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Maja Cigrovski Berković
- Department for Sport and Exercise Medicine, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy; Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Sonja Marinovic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilias Nikas
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Lenka Nosáková
- Clinic of Internal Medicine - Gastroenterology, JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Bratislava, Slovakia
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Eleonora Pellè
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
2
|
Xu J, Shen L, Li J, Zhou Z, Bai C, Li Z, Chi Y, Li E, Yu X, Xu N, Bai Y, Wang X, Yuan X, Liu T, Yin Y, Chen J, Hu H, Li X, Xiu D, Zhang T, Lou W, Ying J, Qin S, Deng Y, Tao M, Cheng Y, Fan S, Luo X, Guo X, Shi MM, Su W. Surufatinib in advanced neuroendocrine tumours: Final overall survival from two randomised, double-blind, placebo-controlled phase 3 studies (SANET-ep and SANET-p). Eur J Cancer 2025; 222:115398. [PMID: 40306120 DOI: 10.1016/j.ejca.2025.115398] [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: 01/07/2025] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND SANET-ep (NCT02588170) and SANET-p (NCT02589821) demonstrated the efficacy and safety of surufatinib versus placebo in patients with advanced extra-pancreatic and pancreatic neuroendocrine tumours (NETs). Here, we present a pooled analysis of final overall survival (OS) from two randomised phase 3 studies. METHODS The SANET studies were randomised, placebo-controlled, double-blind, phase 3 studies in China, comparing the efficacy and safety of oral 300-mg surufatinib (n = 265) versus placebo (n = 133) in patients with unresectable/metastatic, well-differentiated NETs (grade 1/2). After progression of disease or study unblinding, patients receiving placebo crossed over/switched to open-label surufatinib. By pooling the data from the two studies, OS analysis was completed using Kaplan-Meier methodology and a Cox proportional hazards model in the intention-to-treat population. Exploratory analyses were performed using different models to correct the confounding effect introduced by crossover. Long-term safety was assessed. RESULTS At study termination, 69 % of the placebo group had crossed over/switched to surufatinib. Median OS was 50.1 versus 46.8 months for patients initially on surufatinib versus those initially on placebo (stratified hazard ratio [HR] 0.935, 95 % confidence interval [CI] 0.684-1.278; p = 0.6727). After correcting the confounding effect introduced by crossover/switching, the HR ranged from 0.558 to 0.825. Commonly (≥10 %) reported treatment-related adverse events (grade 3/4) included hypertension and proteinuria. CONCLUSION OS of patients initially on surufatinib was not significantly longer versus patients initially on placebo, likely due to the high amount of crossover from placebo to surufatinib. No new safety signals were observed. CLINICAL TRIALS REGISTRATION SANET-ep (NCT02588170) and SANET-p (NCT02589821).
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiwei Zhou
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiping Li
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yihebali Chi
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiuwen Wang
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Chen
- Department of Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Hanguang Hu
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Tao Zhang
- Department of Oncology, Xiehe Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jieer Ying
- Department of Abdominal Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shukui Qin
- Cancer Center of Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, Guangdong, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Songhua Fan
- Department of Clinical Development and Regulatory Affairs, HUTCHMED, Shanghai, China
| | - Xian Luo
- Department of Clinical Development and Regulatory Affairs, HUTCHMED, Shanghai, China
| | - Xiaojun Guo
- Department of Clinical Development and Regulatory Affairs, HUTCHMED, Shanghai, China
| | - Michael M Shi
- Department of Clinical Development and Regulatory Affairs, HUTCHMED, Shanghai, China
| | - Weiguo Su
- Department of Clinical Development and Regulatory Affairs, HUTCHMED, Shanghai, China
| |
Collapse
|
3
|
Price T, Depauw L, Cehic G, Wachter E, Sebben R, Reid J, Neuhaus S, Alawawdeh A, Kirkwood ID, Solanki R, McGregor M, Leopardi L, Rodrigues D, Maddern G. A phase 1 study to assess the safety, tolerability and effectiveness of PV-10 (Rose Bengal Sodium) in neuroendocrine tumours metastatic to the liver. Br J Cancer 2025; 132:888-896. [PMID: 40140696 DOI: 10.1038/s41416-025-02976-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/06/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Metastatic neuroendocrine neoplasms (mNEN) require new treatment options. Intralesional (IL) PV-10 is an autolytic chemotherapy that may elicit an adaptive immune response. METHODS This phase 1 study evaluated IL PV-10 administered percutaneously to hepatic lesions in patients with progressive mNEN. IL PV-10 was injected in a single lesion per treatment cycle. A treatment cycle could be repeated after ≥ 6 weeks if multiple targetable lesions were present. The primary endpoint was safety. RESULTS Twelve patients were enrolled with a median age of 66 years (range 47-79). All patients had progressive disease at enrolment and received prior somatostatin analogues; 10 patients had peptide receptor radionuclide therapy (PRRT) treatment. One lesion was injected per cycle for all 12 patients. Reported grade 3 side effects were photosensitivity (1 patient), face oedema (1 patient), elevated transaminases (1 patient), hypertension (2 patients). Response rate was 42% of injected lesions with patient-level disease control of 84%, PFS 9.4 months and median OS 24.0 months. CONCLUSIONS IL PV-10 elicited no safety concerns. Encouraging evidence of local and systemic disease control was seen in a heavily pre-treated, progressing mNEN population. CLINICAL TRIAL REGISTRATION NUMBER NCT02693067.
Collapse
Affiliation(s)
- Timothy Price
- The Queen Elizabeth Hospital, Woodville, SA, Australia.
- University of Adelaide, Adelaide, SA, Australia.
| | - Laura Depauw
- The Queen Elizabeth Hospital, Woodville, SA, Australia
| | | | - Eric Wachter
- Provectus Biopharmaceuticals, Inc, Knoxville, TN, USA
| | - Ruben Sebben
- The Queen Elizabeth Hospital, Woodville, SA, Australia
| | - Jessica Reid
- The Queen Elizabeth Hospital, Woodville, SA, Australia
| | | | | | - Ian D Kirkwood
- University of Adelaide, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | - Mark McGregor
- The Queen Elizabeth Hospital, Woodville, SA, Australia
| | - Lisa Leopardi
- The Queen Elizabeth Hospital, Woodville, SA, Australia
| | | | - Guy Maddern
- The Queen Elizabeth Hospital, Woodville, SA, Australia
- University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
4
|
Abdeljawaad KAA, Abdelrahman AHM, Sidhom PA, Tallima H, Shoeib T, Mekhemer GAH, Sayed SRM, El-Tayeb MA, Hegazy MEF, Ibrahim MAA. Potential P-glycoprotein (P-gp) inhibitors from SuperDRUG2 database toward reversing multidrug resistance in cancer treatment: Database mining, molecular dynamics, and binding energy estimations. J Mol Graph Model 2025; 137:108997. [PMID: 40043638 DOI: 10.1016/j.jmgm.2025.108997] [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: 05/25/2024] [Revised: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
P-glycoprotein (P-gp) transporter is included in the failure of various carcinoma chemotherapeutics because of the multidrug resistance (MDR) phenomenon, in which the chemotherapeutic drugs are eliminated from target cells. Consequently, inhibiting P-gp transporter function is a prospective strategy for cancer treatment. In the current study, the SuperDRUG2 database containing >4600 pharmaceutical compounds was virtually screened toward the P-gp transporter utilizing the docking predictions. For inhibitors with a docking score lower than -10.5 kcal/mol, molecular dynamics (MD) simulations were performed, accompanied by binding energy evaluations using the MM-GBSA approach. In accordance with the MM-GBSA//100 ns MD, angiotensin amide (SD003508), terlipressin (SD002603), argipressin (SD002535), and lanreotide (SD001365) exhibited potential binding affinities against the P-gp transporter with ΔGbinding < -120.0 kcal/mol. The outstanding consistency of the investigated inhibitors inside the P-gp binding pocket was shown by the post-dynamics analyses. Additionally, MD simulations of the inhibitor-P-gp complexes in a POPC membrane environment were conducted to mimic the physiological conditions. These results demonstrated that angiotensin amide, terlipressin, argipressin, and lanreotide are promising P-gp inhibitors and deserve additional in-vitro/in-vivo studies.
Collapse
Affiliation(s)
- Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt; Academy of Scientific Research & Technology (ASRT), 101 El-Kasr Alaini Street, Cairo, 4262104, Egypt; Frank Laboratory of Neutron Physics, Department of Raman Spectroscopy, Joint Institute for Nuclear Research, Dubna, 141980, Russia.
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt.
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Hatem Tallima
- Department of Chemistry, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt.
| | - Shaban R M Sayed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Mohamed A El-Tayeb
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, 55128, Germany.
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt; Department of Engineering, College of Engineering and Technology, University of Technology and Applied Sciences, Nizwa 611, Oman; School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.
| |
Collapse
|
5
|
Trautwein NF, Mattern S, Hinterleitner M, Reischl G, Schirrmacher R, Steger V, Nadalin S, Nikolaou K, Schwenck J, Singer S, la Fougère C. Histologic Ex Vivo Validation of the [ 18F]SITATE Somatostatin Receptor PET Tracer. J Nucl Med 2025:jnumed.125.269619. [PMID: 40404394 DOI: 10.2967/jnumed.125.269619] [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: 01/31/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Radiolabeled somatostatin analogs (SSAs), such as [68Ga]Ga-DOTA SSAs, have transformed imaging and therapeutic strategies. However, their use is constrained by the high cost of generators and their short half-life. In contrast, [18F]SITATE presents a promising alternative, offering the advantage of a longer half-life than 68Ga, along with the cost-effectiveness of cyclotron-based production. This study evaluated the first histologic ex vivo validation of [18F]SITATE. Methods: This study retrospectively included 47 patients (57% male; mean age, 66.9 ± 14.9 y) with histologically confirmed well-differentiated neuroendocrine neoplasms who underwent [18F]SITATE PET followed by surgery within 4 mo. Lesion uptake was quantified using SUVmean, SUVpeak, SUVmax, and tumor-to-liver ratio (TLR). Histologic somatostatin receptor (SSTR) type 2 expression was determined using histological scores (H-scores), with thresholds defining SSTR scores 1-3. The accuracy of PET imaging for preoperative metastatic detection was evaluated against surgical histology. Results: PET imaging demonstrated a significant correlation between [18F]SITATE uptake (SUVmean and TLR) and SSTR type 2 H-scores (r = 0.618 and 0.622, respectively; P < 0.0001). SSTR score 3 correlated with increased SUVmean and TLR (P < 0.0001). Among 35 patients with primary resection and lymphadenectomy, PET achieved a sensitivity of 73.9% and specificity of 100%. Conclusion: [18F]SITATE PET imaging strongly correlates with histologic SSTR expression, demonstrating utility in staging and guiding therapeutic decisions in neuroendocrine neoplasms. This 18F-labeled tracer shows specificity comparable to historical [68Ga]Ga-DOTA SSA data, whereas an increase in sensitivity for the detection of locoregional metastases appears possible. Further head-to-head comparisons of [18F]SITATE with traditional [68Ga]Ga-DOTA SSA and histologic validation are warranted to optimize its diagnostic accuracy and clinical impact.
Collapse
Affiliation(s)
- Nils F Trautwein
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany;
- ENETS Center of Excellence, University Hospital Tuebingen, Tuebingen, Germany
| | - Sven Mattern
- Department of Pathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Martina Hinterleitner
- ENETS Center of Excellence, University Hospital Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
| | - Gerald Reischl
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, Tuebingen, Germany
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Volker Steger
- Department of Thoracic and Cardiovascular Surgery, German Cardiac Competence Center, University Hospital Tuebingen, Tuebingen, Germany
| | - Silvio Nadalin
- ENETS Center of Excellence, University Hospital Tuebingen, Tuebingen, Germany
- Department of General, Visceral, and Transplant Surgery, University Hospital Tuebingen, Tuebingen, Germany
| | - Konstantin Nikolaou
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany; and
- German Cancer Consortium, German Cancer Research Center, Partner Site Tuebingen, Tuebingen, Germany
| | - Johannes Schwenck
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University Tuebingen, Tuebingen, Germany
| | - Stephan Singer
- ENETS Center of Excellence, University Hospital Tuebingen, Tuebingen, Germany
- Department of Pathology, University Hospital Tuebingen, Tuebingen, Germany
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
| | - Christian la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- ENETS Center of Excellence, University Hospital Tuebingen, Tuebingen, Germany
- DFG Cluster of Excellence 2180 "Image-Guided and Functional Instructed Tumor Therapy," University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium, German Cancer Research Center, Partner Site Tuebingen, Tuebingen, Germany
| |
Collapse
|
6
|
Karimi A, Bogdani C, O'Dwyer E, Siolas D. Emerging innovations in theranostics for pancreatic neuroendocrine tumors. NPJ Precis Oncol 2025; 9:146. [PMID: 40389624 PMCID: PMC12089376 DOI: 10.1038/s41698-025-00938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) often overexpress somatostatin receptor type 2 (SSTR2), making them ideal targets for theranostics, which integrates molecular imaging with targeted radionuclide therapy. 177Lu-DOTATATE significantly extends progression-free survival (22.8 vs. 8.5 months) compared to octreotide LAR. Despite these advances, challenges remain, including treatment resistance and long-term toxicities. In this review, we explore advancements in specialized imaging techniques, rationale combination strategies, and exploring next-generation radiopharmaceuticals.
Collapse
Affiliation(s)
- Anita Karimi
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Christina Bogdani
- New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York, NY, USA
| | - Elisabeth O'Dwyer
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Despina Siolas
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Aggarwal P, Satapathy S, Kaur G, Sood A, Bhadada SK, Walia R, Gupta R, Mittal BR. Safety and Efficacy of Peptide Receptor Radionuclide Therapy in Multiple Endocrine Neoplasia Syndrome: A Single-center Experience. Clin Nucl Med 2025:00003072-990000000-01737. [PMID: 40392141 DOI: 10.1097/rlu.0000000000005891] [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: 01/02/2025] [Accepted: 03/06/2025] [Indexed: 05/22/2025]
Abstract
PURPOSE Peptide receptor radionuclide therapy (PRRT) is recommended and approved in advanced neuroendocrine tumors (NETs). There is a lack of data on the utility of PRRT in multiple endocrine neoplasia (MEN) syndrome. This study explores the utility of PRRT in MEN syndrome patients with inoperable/metastatic NETs. METHODS A single-center retrospective analysis of MEN syndrome patients with advanced NETs referred for PRRT was done. Upto 4 cycles of 177Lu-DOTATATE (5.5-7.4 GBq/cycle) were administered every 8-12 weeks. Treatment-related adverse events were assessed using CTCAE v5. The best response to PRRT was evaluated using RECIST 1.1. Follow-up was done to calculate the progression-free survival (PFS) and overall survival (OS). RESULTS The data of 15 patients with a median age of 36 years (IQR: 32-49) was analyzed. Fourteen patients had MEN-1 syndrome, and 1 had MEN-2 syndrome. Thirteen patients with MEN-1 syndrome had gastroenteropancreatic (GEP) NET, with the multifocal primary as the most common finding (n=7). One patient had thymic carcinoid. All patients had hyperparathyroidism, while 6 had pituitary adenoma. Fifty-three cycles of 177Lu-DOTATATE (range: 1-4) were administered with a median cumulative activity of 27.8 GBq. Twelve patients (80%) showed grade 1/2 adverse events, with leukopenia being the most common. Six patients achieved partial response (43% ORR), 6 showed stable disease (86% DCR), 2 (14%) showed disease progression during PRRT, while 1 patient was lost to follow-up. The median PFS was 32.6 months (95% CI: 14.7-not reached) with an estimated 1- and 5-year PFS rate of 86.2% (95% CI: 68-100) and 46.3% (95% CI: 13-80), and estimated 5-year OS rate of 76.2% (95% CI: 52.1-100). CONCLUSION 177Lu-DOTATATE is a safe and effective treatment option for advanced NETs in MEN syndrome. However, large-sized multicentric prospective studies are required.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rajesh Gupta
- GI Surgery, HPB and Liver Transplantation, PGIMER, Chandigarh, India
| | | |
Collapse
|
8
|
Sahm F, Bertero L, Brandner S, Capper D, Goldbrunner R, Jenkinson MD, Kalamarides M, Lamszus K, Albert NL, Mair MJ, Berghoff AS, Mawrin C, Wirsching HG, Maas SLN, Raleigh DR, Reifenberger G, Schweizer L, Suwala AK, Tabatabai G, Tabouret E, Short S, Wen PY, Weller M, Le Rhun E, Wesseling P, van den Bent M, Preusser M. European Association of Neuro-Oncology guideline on molecular testing of meningiomas for targeted therapy selection. Neuro Oncol 2025; 27:869-883. [PMID: 39577862 PMCID: PMC12083233 DOI: 10.1093/neuonc/noae253] [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] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Indexed: 11/24/2024] Open
Abstract
Meningiomas are the most common primary intracranial tumors of adults. For meningiomas that progress or recur despite surgical resection and radiotherapy, additional treatment options are limited due to a lack of proven efficacy. Meningiomas show recurring molecular aberrations, which may serve as predictive markers for systemic pharmacotherapies with targeted drugs or immunotherapy, radiotherapy, or radioligand therapy. Here, we review the evidence for a predictive role of a wide range of molecular alterations and markers including NF2, AKT1, SMO, SMARCE1, PIK3CA, CDKN2A/B, CDK4/6, TERT, TRAF7, BAP1, KLF4,ARID1/2, SUFU, PD-L1, SSTR2A, PR/ER, mTOR, VEGF(R), PDGFR, as well as homologous recombination deficiency, genomic copy number variations, DNA methylation classes, and combined gene expression profiles. In our assessment based on the established ESMO ESCAT (European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets) evidence-level criteria, no molecular target reached ESCAT I ("ready for clinical use") classification, and only mTOR pathway activation and NF2 alterations reached ESCAT II ("investigational") classification, respectively. Our evaluations may guide targeted therapy selection in clinical practice and clinical trial efforts and highlight areas for which additional research is warranted.
Collapse
Affiliation(s)
- Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg Gemany and CCU Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Luca Bertero
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology and Division of Neuropathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - David Capper
- German Cancer Consortium (DKTK), partner site Berlin and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Roland Goldbrunner
- Department of Neurosurgery, University Hospital Cologne, Cologne, Germany
| | - Michael D Jenkinson
- Department of Neurosurgery, University of Liverpool and Walton Centre, Liverpool, UK
| | - Michel Kalamarides
- Department of Neurosurgery, Pitie-Salpetriere Hospital, AP-HP Sorbonne Université, Paris, France
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | - Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christian Mawrin
- Department of Neuropathology, University Hospital Magdeburg, Magdeburg, Germany
| | - Hans-Georg Wirsching
- Department of Neurology, Brain Tumor Center & Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Sybren L N Maas
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - David R Raleigh
- Departments of Radiation Oncology, Neurological Surgery, and Pathology, University of California, San Francisco, San Francisco, California, USA
| | - Guido Reifenberger
- Institute of Neuropathology, Medical Faculty, Heinrich Heine University and University Hospital Düsseldorf, and German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Leonille Schweizer
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Abigail K Suwala
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg Gemany and CCU Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Ghazaleh Tabatabai
- Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Center for Neuro-Oncology, Comprehensive Cancer Center Tübingen-Stuttgart, DKTK partner site Tübingen, University of Tübingen, Germany
| | - Emeline Tabouret
- Aix-Marseille Universite, APHM, CNRS, INP, Institut Neurophysiopathol, GlioME Team, Plateforme PETRA, CHU Timone, Service de Neurooncologie, Marseille, France
| | - Susan Short
- Department of Oncology, Leeds Institute of Medical Research at St James’s Hospital, Leeds, UK
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Brain Tumor Center & Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emilie Le Rhun
- Department of Medical Oncology and Hematology, Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers/VUmc, Amsterdam, The Netherlands
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Qin X, Wang F, Xie D, Zhou Q, Lin S, Lin W, Li W. Identification of a key peptide cyclase for novel cyclic peptide discovery in Pseudostellaria heterophylla. PLANT COMMUNICATIONS 2025; 6:101315. [PMID: 40083160 DOI: 10.1016/j.xplc.2025.101315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/22/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Orbitides, also known as Caryophyllaceae-type cyclic peptides, from the Traditional Chinese Medicine plant Pseudostellaria heterophylla (Miq.) Pax, exhibit great potential for improving memory and treating diabetes. Orbitides are ribosomally encoded and post-translationally modified peptides; however, the key biosynthetic enzyme mediating this process remains unknown in P. heterophylla. In this study, we investigated the distribution of orbitides in P. heterophylla and mined novel precursor peptide genes and peptide cyclases from multiple omics datasets. The function of PhPCY3, a gene encoding a key tailoring enzyme, was elucidated using transient heterologous expression and virus-induced gene silencing systems. Our findings suggest that PhPCY3 specifically cyclizes linear precursor peptides in planta. Molecular docking and multiple sequence alignment, followed by site-directed mutagenesis, identified N500 and S502 as critical amino acid residues for PhPCY3 function. We identified gene sequences for over 100 precursor peptides and successfully biosynthesized known active orbitides, such as heterophyllin B and pseudostellarin E/F/G. Additionally, four novel orbitides, cyclo-[LDGPPPYF], cyclo-[WGSSTPHT], cyclo-[GLPIGAPWG], and cyclo-[FGDVGPVI], were synthesized using a heterologous expression platform. This study introduces a gene-guided approach for elucidating the biosynthesis pathway and discovering novel orbitides, providing a strategy for mining and biosynthesizing novel orbitides in P. heterophylla and other plants to further investigate their activities.
Collapse
Affiliation(s)
- Xianjin Qin
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Fengjiao Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Dejin Xie
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Qi Zhou
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Sheng Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Juncao Science and Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenxiong Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Juncao Science and Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Wei Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; Bama Yao Autonomous County Rural Revitalization Research Institute, Bama 547500, China.
| |
Collapse
|
10
|
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
|
11
|
Alchaikh Hassan R, Patel A, Dasanu CA. Selecting optimal therapy for advanced non-functional pancreatic neuroendocrine tumors: latest research and therapeutic advances. Expert Opin Pharmacother 2025:1-5. [PMID: 40316435 DOI: 10.1080/14656566.2025.2501142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Affiliation(s)
| | - Arti Patel
- Department of Internal Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, University of California in San Diego Health System, San Diego, CA, USA
| |
Collapse
|
12
|
Aronowitz DI, Hyman K. Emerging therapies and current standards in pulmonary carcinoid management. Curr Opin Pulm Med 2025:00063198-990000000-00242. [PMID: 40265677 DOI: 10.1097/mcp.0000000000001173] [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: 04/24/2025]
Abstract
PURPOSE OF REVIEW Relative to other lung tumors, pulmonary carcinoid tumors are rare and have unique histopathological and clinical features. The purpose of this review is to summarize the presentation and management of pulmonary carcinoid tumors, with a particular focus on recent clinical trials in the management of advanced and metastatic disease. RECENT FINDINGS Surgical resection remains a central tenet in the management of pulmonary carcinoid tumors that are localized and even those that have locoregional spread. However, in recent years, the treatment of pulmonary carcinoid has expanded to include several systemic hormonal and cytotoxic therapies as well as radiation and endobronchial strategies. The decision to initiate any of these therapies as either primary or adjuvant treatment after resection depends upon several factors including tumor stage, disease burden, patient's functional status, and whether they will tolerate surgery. SUMMARY The treatment of pulmonary carcinoid tumors has expanded beyond just surgical resection. Novel regimens of systemic hormonal and cytotoxic therapies as well as radiation and endobronchial intervention should be customized for each patient by a multidisciplinary team of surgeons, medical and radiation oncologists, pulmonologists, and pathologists.
Collapse
Affiliation(s)
- Danielle I Aronowitz
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Northwell Health, New Hyde Park, New York, NY, USA
| | | |
Collapse
|
13
|
Sarker MMR, Fakir S, Kubra KT, Sigdel M, Siejka A, Stepien H, Barabutis N. Lanreotide protects against LPS-induced inflammation in endothelial cells and mouse lungs. Tissue Barriers 2025:2493968. [PMID: 40241488 DOI: 10.1080/21688370.2025.2493968] [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: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025] Open
Abstract
Somatostatin is expressed in various tissues - including the hypothalamus - and strongly suppresses Growth Hormone levels to maintain homeostasis. Synthetic somatostatin analogs are currently used in clinics to treat neuroendocrine tumors and acromegaly. An emerging body of evidence suggests that those synthetic peptides exert anti-inflammatory activities. The present study examines the effect of Lanreotide (LAN) on Lipopolysaccharide (LPS)-triggered injury in endothelial cells and mice. Our findings indicate that LAN effectively mitigates LPS-induced endothelial hyperpermeability, inflammation, and reactive oxygen species (ROS) generation in bovine pulmonary artery endothelial cells (BPAEC) and human lung microvascular endothelial cells (HULEC-5a). A murine model of LPS-induced acute lung injury was also utilized, to examine the effects of LAN in lung edema and inflammation. Our observations suggest that LAN suppresses LPS-induced myosin light chain 2 (MLC2), Cofilin, extracellular signal-regulated kinase 1/2 (ERK1/2), STAT1, STAT3, P38 activation; and lung edema. In conclusion, and based on the aforementioned observations, it is suggested that LAN counteracts experimental LPS-induced injury in endothelial cells and mice.
Collapse
Affiliation(s)
- Md Matiur Rahman Sarker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Saikat Fakir
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Madan Sigdel
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Agnieszka Siejka
- Department of Clinical Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Henryk Stepien
- Department of Immunoendocrinology, Medical University of Lodz, Lodz, Poland
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
14
|
Ji S, Cao L, Gao J, Du Y, Ye Z, Lou X, Liu F, Zhang Y, Xu J, Shi X, Wang H, Li P, Li Y, Chen H, Yang Z, Gao S, Zhang W, Huang D, Ni S, Wei M, Wang F, Wang Y, Ding T, Jing D, Fan G, Gong Z, Lu R, Qin Y, Chen J, Xu X, Wang P, Zhang B, Ding L, Robles AI, Rodriguez H, Chang DK, Hruban RH, Gao D, Gao D, Jin G, Zhou H, Wu J, Yu X. Proteogenomic characterization of non-functional pancreatic neuroendocrine tumors unravels clinically relevant subgroups. Cancer Cell 2025; 43:776-796.e14. [PMID: 40185092 DOI: 10.1016/j.ccell.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
The majority of neuroendocrine neoplasms in pancreas are non-functional pancreatic neuroendocrine tumors (NF-PanNETs), which exhibit a high occurrence of distant metastases with limited therapeutic options. Here, we perform a comprehensive molecular characterization of 108 NF-PanNETs through integrative analysis of genomic, transcriptomic, proteomic, and phosphoproteomic profiles. Proteogenomic analysis provides functional insights into the genomic driver alterations of NF-PanNETs, revealing a potential mediator of MEN1 alterations using Men1-conditional knockout mice. Machine-learning-based modeling uncovers a three-protein signature as an independent prognostic factor, which is validated by an independent external cohort. Proteomic and phosphoproteomic-based stratification identifies four subtypes with distinct molecular characteristics, immune microenvironments, and clinicopathological features. Drug screening using patient-derived tumor organoids identifies cyclin-dependent kinase (CDK) 5 and Calcium Voltage-Gated Channel Subunit Alpha1 D (CACNA1D) as ubiquitous and subtype-specific targets, respectively, with in vivo validation using xenograft models. Together, our proteogenomic analyses illustrate a comprehensive molecular landscape of NF-PanNETs, revealing biological insights and therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Lihua Cao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jing Gao
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Fen Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yehan Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junfeng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Yikai Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Hongxu Chen
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhicheng Yang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Dan Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Miaoyan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Fei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Tian Ding
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Desheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Zhiyun Gong
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Renquan Lu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Jie Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Center for Neuroendocrine Tumors, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Dong Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Daming Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai 200433, China.
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Number 19A Yuquan Road, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Shanghai Institute of Materia Medica-University of Ottawa Joint Research Center in Systems and Personalized Pharmacology, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jianmin Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China; Peking University International Cancer Institute, Peking University, Beijing 100191, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China.
| |
Collapse
|
15
|
Landoni L, Paiella S, Marchetti A, Fontana M, Corvino G, Maistri G, Gronchi F, Cattelani A, Casciani F, Cingarlini S, D'Onofrio M, De Robertis Lombardi R, Malleo G, Salvia R. Somatostatin analogs for resectable pancreatic neuroendocrine tumors in high-risk surgical patients: Data from a single-center cohort. Pancreatology 2025:S1424-3903(25)00066-3. [PMID: 40328552 DOI: 10.1016/j.pan.2025.04.001] [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: 11/17/2024] [Revised: 02/08/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES To evaluate the use of long-acting somatostatin-analogs (SSA) for treating non-advanced, resectable pancreatic neuroendocrine tumors (PNETs) with indications to surgery, in high-risk patients who were not candidates for surgery. METHODS Patients diagnosed with histology-proven, non-advanced G1/low-G2 68Ga-TC/PET-positive PNETs >20 mm who did not undergo surgery due to comorbidities/old age/high-risk surgical profile, which were treated with SSA at a single, high-volume institution were included. The efficacy of SSA was evaluated using the analysis of tumor growth rate (TGR). "Negative TGRTx-TpreSSA" was defined as the first time point after the initiation of SSA when a negative TGRTx-TpreSSA was observed. RESULTS Between 2014 and 2024, 20 patients were treated with long-acting SSA. The median age was 76 (IQR 72-80), and the median ASA score was 3 (IQR 3-3). Fifteen patients (75 %) received Lanreotide, and five (25 %) octreotide acetate (Sandostatin® LAR). The median overall survival was 68.5 months (IQR 60-99). In four patients (20 %), SSA were interrupted when the tumor had shrunk below 2 cm. In one of them, SSA reintroduction was necessary due to disease progression (after 14 months). In two patients (10 %), the treatment was interrupted due to side effects. In one of these, the disease progressed until death at 87 years old. At the last follow-up, sixteen patients were alive, 3 had died (1 death of disease), and 13 were receiving SSA. T2 PNETs presented a significantly faster response to SSA therapy compared to T3 (Median time to "Negative TGRTx-TpreSSA": 6 [95 % CI 5, na] vs 52 [95 % CI 19, na] months, respectively; p = 0.0092) CONCLUSION: In patients at high surgical risk, with localized, resectable, G1 or low-G2, 68Ga-TC/PET-positive PNETs >20 mm, SSA can obtain disease control with a manageable safety profile.
Collapse
Affiliation(s)
- Luca Landoni
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Verona, Italy
| | - Alessio Marchetti
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Michele Fontana
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Gaetano Corvino
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Giulia Maistri
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Federico Gronchi
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Alice Cattelani
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Fabio Casciani
- Unit of Pancreatic Surgery, University of Verona Hospital Trust, Verona, Italy
| | - Sara Cingarlini
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Mirko D'Onofrio
- Department of Radiology, Ospedale G.B. Rossi AOUI Verona, Verona, Italy; Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Riccardo De Robertis Lombardi
- Department of Radiology, Ospedale G.B. Rossi AOUI Verona, Verona, Italy; Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Giuseppe Malleo
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynecology, University of Verona, Verona, Italy
| | - Roberto Salvia
- Pancreatic Surgery Unit, Department of Engineering for Innovation Medicine (DIMI), University of Verona, Verona, Italy.
| |
Collapse
|
16
|
Agoni C, Fernández-Díaz R, Timmons PB, Adelfio A, Gómez H, Shields DC. Molecular Modelling in Bioactive Peptide Discovery and Characterisation. Biomolecules 2025; 15:524. [PMID: 40305228 PMCID: PMC12025251 DOI: 10.3390/biom15040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Molecular modelling is a vital tool in the discovery and characterisation of bioactive peptides, providing insights into their structural properties and interactions with biological targets. Many models predicting bioactive peptide function or structure rely on their intrinsic properties, including the influence of amino acid composition, sequence, and chain length, which impact stability, folding, aggregation, and target interaction. Homology modelling predicts peptide structures based on known templates. Peptide-protein interactions can be explored using molecular docking techniques, but there are challenges related to the inherent flexibility of peptides, which can be addressed by more computationally intensive approaches that consider their movement over time, called molecular dynamics (MD). Virtual screening of many peptides, usually against a single target, enables rapid identification of potential bioactive peptides from large libraries, typically using docking approaches. The integration of artificial intelligence (AI) has transformed peptide discovery by leveraging large amounts of data. AlphaFold is a general protein structure prediction tool based on deep learning that has greatly improved the predictions of peptide conformations and interactions, in addition to providing estimates of model accuracy at each residue which greatly guide interpretation. Peptide function and structure prediction are being further enhanced using Protein Language Models (PLMs), which are large deep-learning-derived statistical models that learn computer representations useful to identify fundamental patterns of proteins. Recent methodological developments are discussed in the context of canonical peptides, as well as those with modifications and cyclisations. In designing potential peptide therapeutics, the main outstanding challenge for these methods is the incorporation of diverse non-canonical amino acids and cyclisations.
Collapse
Affiliation(s)
- Clement Agoni
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, D04 C1P Dublin, Ireland
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Raúl Fernández-Díaz
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- IBM Research, D15 HN66 Dublin, Ireland
| | | | - Alessandro Adelfio
- Nuritas Ltd., Joshua Dawson House, D02 RY95 Dublin, Ireland; (P.B.T.); (A.A.); (H.G.)
| | - Hansel Gómez
- Nuritas Ltd., Joshua Dawson House, D02 RY95 Dublin, Ireland; (P.B.T.); (A.A.); (H.G.)
| | - Denis C. Shields
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland;
- Conway Institute of Biomolecular and Biomedical Science, University College Dublin, D04 C1P Dublin, Ireland
| |
Collapse
|
17
|
Kiesewetter B, Pflüger FF, Melhorn P, Mazal P, Raderer M. Long-term experience with octreotide and lanreotide for the treatment of gastroenteropancreatic neuroendocrine tumors. Clin Transl Oncol 2025; 27:1642-1652. [PMID: 39316250 PMCID: PMC12000220 DOI: 10.1007/s12094-024-03732-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] [Received: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The somatostatin analogs (SSA) octreotide and lanreotide are a mainstay in the treatment of neuroendocrine tumors (NET). The two pivotal trials differed considerably in terms of patient characteristics and are not directly comparable. Further comparative data are lacking. METHODS This retrospective chart review study included patients with gastroenteropancreatic NET grade 1 or 2 who were treated with octreotide LAR or lanreotide autogel. The main aim was to compare the two SSA based on progression-free survival (PFS) and overall survival (OS) from treatment start. RESULTS In total, 129 patients were analyzed, 60% (n = 77) had a small intestinal NET and 31% (n = 40) a pancreatic NET. Histologically, 34% (n = 44) had NET G1, 55% (n = 71) a NET G2, and 11% (n = 14) a NET G1/G2 unclassified. Lanreotide was used in 90 patients (70%) and octreotide in 39 patients (30%). Overall, the median PFS was 32.2 months (95% CI 23.0-42.9 months). No PFS difference (p = 0.8) was observed between lanreotide (29.8 months, 95% CI 18.7-48.5 months) and octreotide (36.0 months, 95% CI 23.2-68.2 months). Median OS from treatment start was calculated at 93.5 months (95% CI 71.1-132.9 months). Again, the median OS following lanreotide (113.4 months, 95% CI 62.3-NA months) or after octreotide (90.3 months, 95% CI 71.1-NA months) did not differ significantly (p > 0.9). CONCLUSIONS Our long-term experience with octreotide and lanreotide in NET did not reveal differences in antitumor effectiveness. This is consistent with previous reports and might suggest that both SSA can be used interchangeably if needed.
Collapse
Affiliation(s)
- Barbara Kiesewetter
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
| | - Friedrich Franz Pflüger
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Philipp Melhorn
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Peter Mazal
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Markus Raderer
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| |
Collapse
|
18
|
Murase Y, Esaki M, Mizui T, Takamoto T, Nara S, Ban D, Hiraoka N, Shimada K. Optimal lymph node dissection area for pancreatic neuroendocrine neoplasms by tumor location, size, and grade. Surgery 2025; 180:109029. [PMID: 39740608 DOI: 10.1016/j.surg.2024.109029] [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: 07/18/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Lymph node dissection is required for many pancreatic neuroendocrine neoplasms. However, the need for such dissection has rarely been examined in detail by the tumor size, tumor location, or World Health Organization grading. The objective is to determine which characteristics of pancreatic neuroendocrine neoplasms require lymph node dissection, and to what extent lymph node dissection should be performed. METHODS A retrospective review was performed of patients who had undergone pancreatectomy with lymph node dissection between 2000 and 2022. Frequency of lymph node metastases stratified by tumor size and grade, and efficacy index (calculated by multiplying the frequency of metastasis to the lymph node station by the 5-year overall survival rate of patients with metastasis to that station) for each lymph node station were evaluated. RESULTS Among 130 patients, 29 (22.3%) had lymph node metastases. Tumors larger than 2 cm had a 33.3% lymph node metastasis rate, whereas grade 1 and grade 2 neuroendocrine tumors had rates of 2.4% and 31.8%, respectively. In grade 1 neuroendocrine tumor, for pancreatic head tumor, only peripancreatic head lymph node had an efficacy index of 5.0. Pancreatic body and tail tumor had no lymph node metastases. In grade 2 neuroendocrine tumor, for pancreatic head tumor, peripancreatic and superior mesenteric artery lymph nodes had efficacy indexes of 29.2 and 14.3, respectively. For pancreatic body tumor, peridistal pancreatic lymph node had an efficacy index of 27.3, whereas for pancreatic tail tumor, peridistal pancreatic lymph node and splenic hilum lymph node had an efficacy index of 27.8 and 7.1, respectively. CONCLUSION The optimal extent of lymph node dissection for pancreatic neuroendocrine neoplasms should be determined by considering both the tumor size and grade for each tumor location.
Collapse
Affiliation(s)
- Yoshiki Murase
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Minoru Esaki
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan.
| | - Takahiro Mizui
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Takeshi Takamoto
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Nara
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Nobuyoshi Hiraoka
- Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazuaki Shimada
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Sahu A, Patlas M, Jajodia A. The radiologic spectrum of neuroendocrine tumors in emergent care. Rev Endocr Metab Disord 2025; 26:175-186. [PMID: 39745542 DOI: 10.1007/s11154-024-09940-7] [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: 12/29/2024] [Indexed: 03/19/2025]
Abstract
Neuroendocrine tumors (NETs) are a diverse group of neoplasms whose prevalence is increasing globally, primarily due to advancements in diagnostic techniques. NETs arise from cells of the diffuse endocrine system and can occur in various locations, with the gastrointestinal tract being the most common. Their diverse clinical presentations, which range from asymptomatic to severe hormone-induced syndromes, pose significant diagnostic challenges. In emergency care, prompt recognition and management of complications such as bowel obstruction, ischemic events, hormonal crises, and metastases are critical. This review discusses the radiologic spectrum of NETs in emergent care, emphasizing the role of imaging in timely diagnosis and intervention.
Collapse
Affiliation(s)
- Asutosh Sahu
- Emergency, Trauma & Acute Care Radiology St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Michael Patlas
- Division of Abdominal Imaging, Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada
| | - Ankush Jajodia
- Division of Abdominal Imaging, Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
Campos Ramírez SE, Andrés García A, Blanco Abad C, Gomila Pons P, Gómez Mugarza P, Ruffini Egea SE, Gallart Caballero L, Polo Marques E, Alonso Orduña V. Evaluation of the Real-Life Efficacy and Safety of the Treatment with Lutetium-177 Dotatate for Metastatic Neuroendocrine Tumors. J Clin Med 2025; 14:2384. [PMID: 40217834 PMCID: PMC11989597 DOI: 10.3390/jcm14072384] [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: 03/02/2025] [Revised: 03/20/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Therapy using lutetium-177 dotatate (177LU) was approved in Europe for the treatment of advanced neuroendocrine tumors (NETs) in 2017. Since then, it has become part of the strategies in the treatment of NETs, making it now possible to evaluate real-life results. Research Design and Methods: Single-arm, retrospective, multicenter, cohort study of all the patients with metastatic NETs treated with 177LU (four cycles of 200 mCi every 8 weeks) in the two medical centers dedicated to the treatment of NETs from the region of Aragón, Spain, from 2017 to 2024. Descriptive analysis of demographic characteristics, efficacy, and survival analysis were performed using the statistics software Jamovi 2.6.14. Results: Sixty-eight patients were included. The majority were male, and the most frequent primary location was the pancreas. The ORR was 30.9%. The DCR was 88%. The median OS was 47.4 months [95% CI, 25.6-NE]. The median PFS was 26.1 months [95% CI, 18.5-68.3]. High-grade tumors, multiple previous treatments, and pancreatic location presented worse OS. In total, 42.6% presented any grade adverse event (17.2% hematologic, 30.9% GI symptoms). Conclusions: The efficacy of 177LU in our study is like that observed in similar studies. Acceptable tolerance has been shown. Pancreatic tumors, previous treatments, and higher grades demonstrated worse outcomes. The new research line must consider the use of treatment with 177LU in earlier lines for metastatic disease as well as its possible use in local or locally advanced disease.
Collapse
Affiliation(s)
| | - Alejandro Andrés García
- Department of Nuclear Medicine, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | - Carmen Blanco Abad
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Paula Gomila Pons
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Pablo Gómez Mugarza
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | | | - Luis Gallart Caballero
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Eduardo Polo Marques
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Vicente Alonso Orduña
- Department of Medical Oncology, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| |
Collapse
|
21
|
Rościszewska A, Tokarska K, Kośny A, Karp P, Leja W, Żebrowska A. The Importance and Challenges of Early Diagnosis of Paraneoplastic Skin Syndromes in Cancer Detection-A Review. Cancers (Basel) 2025; 17:1053. [PMID: 40227497 PMCID: PMC11988160 DOI: 10.3390/cancers17071053] [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: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Skin paraneoplastic syndromes (SPNSs) are a group of disorders that arise as a consequence of cancer but are not directly related to the tumor mass itself. This review aims to provide a comprehensive overview of these syndromes, encompassing their pathophysiology, clinical features, diagnostic approaches, differential diagnosis, and management strategies. These syndromes, which include conditions such as Bazex syndrome, acanthosis nigricans, dermatomyositis, and necrolytic migratory erythema often manifest prior to or concurrently with a cancer diagnosis, serving as potential early warning signs of underlying malignancies. This review delves into the spectrum of SPNSs and their associations with specific cancer types. Special emphasis is placed on the critical role of dermatologists and oncologists in identifying these skin manifestations as potential markers of malignancy. By raising awareness of SPNSs, this paper highlights the pivotal importance of prompt recognition and intervention in reducing cancer-related mortality.
Collapse
Affiliation(s)
| | | | | | | | | | - Agnieszka Żebrowska
- Department of Dermatology and Venereology, Medical University of Lodz, Hallera 1, 90-647 Lodz, Poland; (A.R.); (K.T.); (A.K.); (P.K.); (W.L.)
| |
Collapse
|
22
|
Leigh J, Ahmed A, Aubin F, Berry S, Boucher M, Campeau MP, Colwell B, Connors S, Corbett J, Dadwal S, Dudani S, Elimova E, Falkson C, Galvis L, Goel R, Gotfrit J, Hyde A, Febbraro M, Laidley DT, Locke G, Mahmud A, Baccili Cury Megid T, Michael J, Nair VJ, Quigley S, Ramjeesingh R, Samimi S, Seal M, Snow S, Spadafora S, Stuckless T, Wilson B, Asmis T, Goodwin R, Vickers M. Eastern Canadian Gastrointestinal Cancer Consensus Conference 2024. Curr Oncol 2025; 32:175. [PMID: 40136379 PMCID: PMC11941643 DOI: 10.3390/curroncol32030175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
The Eastern Canadian Gastrointestinal Cancer Consensus Conference was an annual meeting that was held in St. John's, Newfoundland and Labrador, from 26 to 28 September 2024. This included experts in medical oncology, radiation oncology, surgical oncology, nuclear medicine, and general practitioners in oncology (GPO) from across the eastern Canadian provinces who are involved in the management of patients with gastrointestinal malignancies. This consensus statement generated by the conference addresses multiple topics, including the management of localized rectal cancer, liver-limited colorectal cancer, systemic therapy for advanced biliary tract cancers, radioligand therapy for gastroenteropancreatic neuroendocrine tumors (GEP-NETs), systemic therapy for pancreatic and midgut well-differentiated NETs, and systemic therapy for HER2-positive gastroesophageal cancers.
Collapse
Affiliation(s)
| | - Arwa Ahmed
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Francine Aubin
- Centre Hospitalier de l’Universite de Montreal, Montreal, QC H2X 3E4, Canada
| | - Scott Berry
- Trillium Health Partners, Mississauga, ON L5A 4G1, Canada
| | - Melanie Boucher
- Prince Edward Island Cancer Treatment Center, Charlottetown, PE C1A 8T5, Canada
| | | | - Bruce Colwell
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | | | - Jessica Corbett
- Prince Edward Island Cancer Treatment Center, Charlottetown, PE C1A 8T5, Canada
| | | | - Shaan Dudani
- William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Elena Elimova
- Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada
| | - Conrad Falkson
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | - Luisa Galvis
- Horizon Health Network, Fredericton, NB E3B 4R3, Canada
| | - Rakesh Goel
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Joanna Gotfrit
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Angela Hyde
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Michela Febbraro
- Algoma District Cancer Program, Sault Ste. Marie, ON P6B 0A8, Canada
| | | | - Gordon Locke
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Aamer Mahmud
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | | | - James Michael
- Saint John Regional Hospital Oncology Center, Saint John, NB E2L 4L2, Canada
| | - Vimoj J. Nair
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Stephen Quigley
- Health Sciences Center-Eastern Health, St. John’s, NL A1B 3V6, Canada
| | - Ravi Ramjeesingh
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | - Setareh Samimi
- Hopital du Sacre-Coeur de Montreal, Montreal, QC H4J 1C5, Canada
| | - Melanie Seal
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Stephanie Snow
- Queen Elizabeth II Health Sciences Center, Halifax, NS B3H 3A7, Canada
| | - Silvana Spadafora
- Algoma District Cancer Program, Sault Ste. Marie, ON P6B 0A8, Canada
| | - Teri Stuckless
- Dr. H. Bliss Murphy Cancer Center, St. John’s, NL A1B 3X5, Canada
| | - Brooke Wilson
- Kingston Health Sciences Center, Kingston, ON K7L 2V7, Canada
| | - Timothy Asmis
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Rachel Goodwin
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| | - Michael Vickers
- The Ottawa Hospital Cancer Centre, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
23
|
Fernandez-Cuesta L, Alcala N, Mathian E, Derks J, Thirlwell C, Dayton T, Marinoni I, Perren A, Walter T, Foll M. Basic science and translational implications of current knowledge on neuroendocrine tumors. J Clin Invest 2025; 135:e186702. [PMID: 40026252 PMCID: PMC11870734 DOI: 10.1172/jci186702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Neuroendocrine tumors (NETs) are a diverse group of malignancies that can occur in various organs, with a notable prevalence in the lungs and gastrointestinal tract, which are the focus of this Review. Although NETs are rare in individual organs, their incidence has increased over recent decades, highlighting the urgent need for current classification systems to evolve by incorporating recent advances in the understanding of NET biology. Several omics studies have revealed molecular subtypes, which, when integrated into existing classification frameworks, may provide more clinically relevant insights for patients with NETs. This Review examines recent progress in elucidating the biology of NETs, with a particular emphasis on the tumor microenvironment and cells of origin. The existence of different cells of origin, which may contribute to distinct molecular groups, along with profiles of immune infiltration - despite being generally low - could explain the emergence of more aggressive cases and the potential for metastatic progression. Given the molecular heterogeneity of NETs and the diversity of their microenvironments and different cells of origin, there is an urgent need to develop morphomolecular classification systems. Such systems would make it possible to better characterize tumor progression, identify new therapeutic targets, and, ultimately, guide the development of personalized therapies.
Collapse
Affiliation(s)
- Lynnette Fernandez-Cuesta
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Nicolas Alcala
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Emilie Mathian
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Jules Derks
- Department of Pulmonary Medicine, Erasmus MC Cancer institute, University Medical Center, Rotterdam, Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Talya Dayton
- European Molecular Biology Laboratory Barcelona, Tissue Biology and Disease Modeling, Barcelona, Spain
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Thomas Walter
- Service d’Oncologie Médicale, Groupement Hospitalier Centre, Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France
| | - Matthieu Foll
- Computational Cancer Genomics Team, Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| |
Collapse
|
24
|
Rodrigues A, Henrique R, Jerónimo C, Araújo A. Management of typical and atypical metastatic lung carcinoids: present and future perspectives. Clin Transl Oncol 2025; 27:816-823. [PMID: 39110397 DOI: 10.1007/s12094-024-03607-0] [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: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 09/21/2024]
Abstract
Lung carcinoids are rare tumors representing 1-2% of all invasive lung malignancies. They include typical and atypical carcinoids, whose distinction is made based on the mitotic index and presence or absence of necrosis. The 10-year overall survival for stage IV typical carcinoid is 47% and 18% for atypical carcinoid, reflecting the indolent growth of these tumors. There are limited approved treatment options for them and most of the evidence comes from retrospective analyses, single-arm trials, subgroup analysis of phase II/III trials for metastatic neuroendocrine tumors and extrapolation of data from phase III trials for gastroenteropancreatic neuroendocrine tumors. Management of metastatic lung carcinoids requires a multidisciplinary standardized approach in specialized centers. Treatment should have a dual objective, control of tumor growth and control of symptoms related to hypersecretion syndromes, aiming to improve quality of life and survival. In the continuum of treatment disease, locoregional treatment options need to be considered in parallel with systemic treatments. In this paper, we review the present treatment options and their rational and we give an insight into future alternatives.
Collapse
Affiliation(s)
- Ana Rodrigues
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.
| | - Rui Henrique
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira no 228, 4050-313, Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Carmen Jerónimo
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira no 228, 4050-313, Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - António Araújo
- Department of Medical Oncology, ULS de Santo António, Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
25
|
Chen JS, Bai LY, Cheng HH, Chan SL, Zou JY, Shi X, Houchard A, Truong-Thanh XM, Chen MH. Real-World Study of Lanreotide Autogel in Routine Practice in Patients with Gastroenteropancreatic Neuroendocrine Tumors (GEP-NETs) in Hong Kong and Taiwan. Oncol Ther 2025; 13:69-83. [PMID: 39215958 PMCID: PMC11880440 DOI: 10.1007/s40487-024-00302-6] [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: 05/30/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION There is a lack of data on the efficacy, effectiveness, and safety of lanreotide autogel in patients with gastroenteropancreatic neuroendocrine tumors (GEP-NETs) of Chinese ethnicity. This noninterventional, retrospective study evaluated the effectiveness and safety of lanreotide autogel in patients of Chinese ethnicity with GEP-NETs in clinical practice. METHODS Patients' charts were abstracted from five hospitals in Hong Kong and Taiwan (July-September 2021), where lanreotide autogel is approved for treating GEP-NETs. Included patients were adults with unresectable, metastatic, or locally advanced GEP-NETs who received a first injection (index) of lanreotide autogel 120 mg between 01 January 2017 and 30 June 2020 (planned sample size: N = 30). Follow-up ran from index to a maximum of 48 (± 4) weeks or until disease progression, start of new antitumor treatment, or death. The primary endpoint was progression-free survival (PFS) rate at week 48 (±4), and secondary endpoints included PFS rate at week 24 (±4), estimated using Kaplan-Meier analyses. All analyses were descriptive. RESULTS Of 27 patients enrolled, 22 (81.5%) had 48 weeks of follow-up. Tumors of pancreatic origin were the most common (73.9%). PFS rate was 0.96 (95% confidence interval: 0.72 - 0.99) at 24 weeks and 0.82 (0.53-0.94) at 48 weeks. Overall, 74.1% patients experienced ≥ 1 treatment-emergent adverse event; none were serious. No deaths were reported. CONCLUSIONS Lanreotide autogel was well tolerated and showed good tumor control rate in a real-world setting. These findings align with results from previous studies in Caucasian, Japanese, and Korean patients, thus supporting lanreotide autogel for treating patients with GEP-NETs of Chinese ethnicity.
Collapse
Affiliation(s)
- Jen-Shi Chen
- Department of Hematology-Oncology, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan City, Taiwan
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | - Stephen Lam Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | - Ming-Huang Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei City, 11217, Taiwan.
| |
Collapse
|
26
|
Sahithi PVL, Nandi VPKR, Kandagaddala Y, Mulagapaka V, Onteddu LP, Onteddu NK. Gastrinoma: A Case of Chronic Diarrhea. Cureus 2025; 17:e81253. [PMID: 40291307 PMCID: PMC12031656 DOI: 10.7759/cureus.81253] [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] [Received: 02/08/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Gastrinoma is a rare neuroendocrine tumor characterized by the hypersecretion of gastrin. Early diagnosis is challenging due to its rarity, and patients present to the outpatient department with nonspecific complaints such as diarrhea, weight loss, and fatigue, which often lead to a broad differential diagnosis. This report presents the case of a 59-year-old female who has come to the internal medicine department for a workup of chronic diarrhea, low-grade fever, 8kg weight loss, and severe reflux of eight months duration, not responding to empiric therapy. After multiple hospital visits and evaluations, she was found to have duodenal gastrinoma. This case report aims to create awareness and underscores the importance of considering neuroendocrine tumors as an initial differential diagnosis for patients with nonspecific or unexplained gastrointestinal symptoms to facilitate early diagnosis.
Collapse
Affiliation(s)
| | | | | | - Vinusha Mulagapaka
- Internal Medicine, P.E.S. Institute of Medical Sciences and Research, Kuppam, IND
| | | | - Nirmal K Onteddu
- Internal Medicine, University of Florida College of Medicine, Jacksonville, USA
| |
Collapse
|
27
|
Ranallo N, Roncadori A, Gentili N, Balzi W, Altini M, Ghini V, Maltoni R, Andalò A, Cavallucci M, Sansovini M, Fausti V, Montella MT, Massa I, Danesi V. Treatments and Outcomes in Neuroendocrine Patients Treated with Long-Acting Somatostatin Analogues: An Italian Real-World Propensity Score-Matched Cohort Study. Biomedicines 2025; 13:515. [PMID: 40002928 PMCID: PMC11852996 DOI: 10.3390/biomedicines13020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Objectives: The aim of this study was to investigate the treatment patterns and outcomes in two propensity score-matched cohorts of patients with neuroendocrine tumours (NETs) treated with first-line somatostatin analogue (SSA). Methods: Metastatic NET patients treated with first-line SSA (2009-2022) were retrospectively examined. First-line lanreotide vs. octreotide cohorts were matched 1:1 by propensity scores for demographics, tumour characteristics, and diagnosis year. Progression-free survival (PFS) and overall survival (OS) were analysed using Kaplan-Meier analysis and the Cox proportional hazards model. Results: Among 441 patients, 310 were matched (155 in both the octreotide and lanreotide groups). First-line SSA was monotherapy (63.5%) or combination with other medications (36.5%). A total of 77% of second-line patients (188/244) maintained their initial SSA medication in combination with other therapies. Radioligand therapy with lanreotide (N = 72; 29.5%) or octreotide (N = 70; 28.7%) was the most common second-line treatment. First-line lanreotide and octreotide cohorts had similar median PFS (15.5; 95% CI: 13.6-19.1 vs. 14.0; 95% CI: 12.0-15.8 months), despite octreotide having a 36% higher likelihood of moving to the second line than lanreotide (95% CI: 1.05-1.76, p = 0.018). Multiple metastases (HR = 1.45; p = 0.004, 95% CI: 1.13-1.87) and Ki-67 > 20% (HR = 2.34; p < 0.001, 95% CI: 1.43-3.83) were significantly associated with the worst PFS. First-line lanreotide patients had a median OS of 10.4 years (95% CI: 7.5-NA) and octreotide 9.2 years (95% CI: 7.3-NA) (p = 0.537). Bone metastases increased death risk by 91% (p = 0.014; 95% CI: 1.14-3.20). Conclusions: SSA monotherapy is the main first-line treatment and most subsequent treatments include SSA with additional medications. Cohorts had similar PFS/OS, but octreotide demonstrated a 36% significantly higher likelihood of moving to the second-line treatment.
Collapse
Affiliation(s)
- Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (V.G.); (V.F.)
| | - Andrea Roncadori
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| | - Nicola Gentili
- Data Unit, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.G.); (M.C.)
| | - William Balzi
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| | - Mattia Altini
- Assistenza Ospedaliera Regione Emilia-Romagna, 40127 Bologna, Italy;
| | - Virginia Ghini
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (V.G.); (V.F.)
| | - Roberta Maltoni
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| | - Alice Andalò
- Data Unit, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.G.); (M.C.)
| | - Martina Cavallucci
- Data Unit, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.G.); (M.C.)
| | - Maddalena Sansovini
- Nuclear Medicine Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valentina Fausti
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (V.G.); (V.F.)
| | - Maria Teresa Montella
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| | - Ilaria Massa
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| | - Valentina Danesi
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.R.); (W.B.); (R.M.); (M.T.M.); (I.M.); (V.D.)
| |
Collapse
|
28
|
Chan JA, Geyer S, Zemla T, Knopp MV, Behr S, Pulsipher S, Ou FS, Dueck AC, Acoba J, Shergill A, Wolin EM, Halfdanarson TR, Konda B, Trikalinos NA, Tawfik B, Raj N, Shaheen S, Vijayvergia N, Dasari A, Strosberg JR, Kohn EC, Kulke MH, O’Reilly EM, Meyerhardt JA. Phase 3 Trial of Cabozantinib to Treat Advanced Neuroendocrine Tumors. N Engl J Med 2025; 392:653-665. [PMID: 39282913 PMCID: PMC11821447 DOI: 10.1056/nejmoa2403991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
BACKGROUND Treatment options for patients with advanced neuroendocrine tumors are limited. The efficacy of cabozantinib in the treatment of previously treated, progressive extrapancreatic or pancreatic neuroendocrine tumors is unclear. METHODS We enrolled two independent cohorts of patients - those with extrapancreatic neuroendocrine tumors and those with pancreatic neuroendocrine tumors - who had received peptide receptor radionuclide therapy or targeted therapy or both. Patients were randomly assigned in a 2:1 ratio to receive cabozantinib at a dose of 60 mg daily or placebo. The primary end point was progression-free survival as assessed by blinded independent central review. Key secondary end points included objective response, overall survival, and safety. RESULTS In the cohort of 203 patients with extrapancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 8.4 months, as compared with 3.9 months with placebo (stratified hazard ratio for progression or death, 0.38; 95% confidence interval [CI], 0.25 to 0.59; P<0.001). In the cohort of 95 patients with pancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 13.8 months, as compared with 4.4 months with placebo (stratified hazard ratio, 0.23; 95% CI, 0.12 to 0.42; P<0.001). The incidence of confirmed objective response with cabozantinib was 5% and 19% among patients with extrapancreatic and pancreatic neuroendocrine tumors, respectively, as compared with 0% with placebo. Grade 3 or higher adverse events were noted in 62 to 65% of the patients treated with cabozantinib, as compared with 23 to 27% of the patients who received placebo. Common treatment-related adverse events of grade 3 or higher included hypertension, fatigue, diarrhea, and thromboembolic events. CONCLUSIONS Cabozantinib, as compared with placebo, significantly improved progression-free survival in patients with previously treated, progressive advanced extrapancreatic or pancreatic neuroendocrine tumors. Adverse events were consistent with the known safety profile of cabozantinib. (Funded by the National Cancer Institute and others; CABINET ClinicalTrials.gov number, NCT03375320.).
Collapse
Affiliation(s)
| | - Susan Geyer
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Tyler Zemla
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Michael V. Knopp
- Wright Center of Innovation & IROC, University of Cincinnati, Cincinnati, OH
| | - Spencer Behr
- University of California, San Francisco, San Francisco, CA
| | - Sydney Pulsipher
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Amylou C. Dueck
- Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ
| | - Jared Acoba
- University of Hawaii Cancer Center, Honolulu, HI
| | - Ardaman Shergill
- Alliance Protocol Operations Office, University of Chicago, Chicago, IL
| | | | | | - Bhavana Konda
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Bernard Tawfik
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM
| | - Nitya Raj
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Haimi M, Yang JW, Kremer R. Refractory hypercalcemia caused by parathyroid-hormone-related peptide secretion from a metastatic pancreatic neuroendocrine tumor: a case report. J Med Case Rep 2025; 19:54. [PMID: 39924477 PMCID: PMC11809048 DOI: 10.1186/s13256-025-05074-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/13/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND The parathyroid-hormone-related peptide has been shown in earlier studies to be secreted by pancreatic neuroendocrine tumors, although its secretion by gastroenteropancreatic neuroendocrine tumors is very rare. In contrast, a number of solid tumors, such as lung cancer and renal cell carcinoma, have frequently been shown to secrete parathyroid-hormone-related peptide. CASE PRESENTATION We describe a case report of a 53-year-old Canadian white patient with refractory parathyroid-hormone-related-peptide-mediated hypercalcemia associated with metastatic pancreatic neuroendocrine tumors and review the available research. Our patient had severe hypercalcemia initially refractory to treatment. Computed tomography scan of the abdomen revealed a pancreatic lesion and multiple hepatic metastases. A liver biopsy confirmed metastatic pancreatic neuroendocrine tumor expressing parathyroid-hormone-related peptide. Circulating parathyroid-hormone-related peptide levels were at the upper limit of normal preoperatively and decreased sharply postoperatively following debulking of the tumor. Blood calcium levels eventually normalized on long-term administration of the somatostatin analog lanreotide in combination with denosumab. CONCLUSIONS We describe a case with parathyroid-hormone-related-peptide-mediated hypercalcemia in a pancreatic neuroendocrine tumor (parathyroid-hormone-related peptide tumor). Refractory hypercalcemia was likely the result of parathyroid-hormone-related peptide overproduction by the tumor and resolved following normalization of parathyroid-hormone-related peptide levels.
Collapse
Affiliation(s)
- Motti Haimi
- Meuhedet Health Services, Northern District, Israel.
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3109601, Haifa, Israel.
- Health Systems Management Department, The Max Stern Yezreel Valley College, Yezreel Valley, Israel.
| | - Ji Wei Yang
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Richard Kremer
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
30
|
Massironi S, Albertelli M, Hasballa I, Paravani P, Ferone D, Faggiano A, Danese S. "Cold" Somatostatin Analogs in Neuroendocrine Neoplasms: Decoding Mechanisms, Overcoming Resistance, and Shaping the Future of Therapy. Cells 2025; 14:245. [PMID: 39996718 PMCID: PMC11854070 DOI: 10.3390/cells14040245] [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] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Neuroendocrine neoplasms (NENs) represent a heterogeneous group of tumors that pose significant therapeutic challenges due to their potential for progression, metastasis, and hormonal syndromes. Somatostatin analogs (SSAs) have emerged as a cornerstone in NEN treatment, offering both antisecretory and antiproliferative effects by targeting somatostatin receptors (SSTRs). Despite their proven efficacy, intrinsic and acquired resistance mechanisms, including receptor downregulation, tumor heterogeneity, and microenvironmental influences, limit their long-term effectiveness. Recent advances, including high-dose SSA regimens and novel formulations, have aimed to optimize their therapeutic utility and address these limitations. Body of the review. This review explores the cellular and molecular mechanisms underlying the antitumor effects of SSAs, including receptor-mediated signaling pathways, cell cycle arrest, apoptosis induction, and antiangiogenesis. The role of SSAs in combination therapies with mTOR inhibitors and peptide receptor radionuclide therapy (PRRT) is analyzed, emphasizing their synergistic potential. Key clinical trials, such as RADIANT-2, EVERLAR, and NETTER-1, support the efficacy of these approaches, demonstrating improved outcomes when SSAs are combined with targeted agents or radiolabeled therapies. Emerging strategies include high-dose SSA regimens, particularly in progressive cases with low Ki67 indices. Finally, novel formulations, including oral octreotide, paltusotine, and subcutaneous depot formulations like CAM2029, offer improved pharmacokinetics, bioavailability, and patient adherence. Ongoing clinical trials, including SORENTO, further evaluate their efficacy and safety profiles. CONCLUSIONS This paper provides a comprehensive analysis of the cellular and molecular mechanisms of SSAs. SSAs remain integral to the management of NENs, providing effective tumor stabilization and symptom control. However, resistance mechanisms and tumor heterogeneity necessitate innovative approaches, including high-dose regimens, combination strategies, and next-generation formulations. Future research should focus on refining these strategies to optimize patient outcomes, enhance long-term efficacy, and expand the therapeutic landscape for NENs.
Collapse
Affiliation(s)
- Sara Massironi
- Faculty of Medicine and Surgery, Vita e Salute San Raffaele University, Via Olgettina, 20132 Milan, Italy;
- Gastroenterology Unit, Istituti Ospedalieri Bergamaschi, 24046 Bergamo, Italy
| | - Manuela Albertelli
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Iderina Hasballa
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
| | - Piero Paravani
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (P.P.); (A.F.)
| | - Diego Ferone
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
| | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (P.P.); (A.F.)
| | - Silvio Danese
- Faculty of Medicine and Surgery, Vita e Salute San Raffaele University, Via Olgettina, 20132 Milan, Italy;
- Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| |
Collapse
|
31
|
del Olmo-García M, Hernandez-Rienda L, Garcia-Carbonero R, Hernando J, Custodio A, Anton-Pascual B, Gomez M, Palma Milla S, Suarez L, Bellver M, Alonso V, Serrano R, Valdés N, Melian M, Febrero B, Sampedro-Nuñez MA, Biarnes J, Díaz-Pérez JÁ, Molina-Cerrillo J, Lopez C, Martínez Olmos MÁ, Merino-Torres JF, Capdevila J, Argente Pla M. Nutritional status and quality of life of patients with advanced gastroenteropancreatic neuroendocrine neoplasms in Spain: the NUTRIGETNE (GETNE-S2109) study. Oncologist 2025; 30:oyae343. [PMID: 39998905 PMCID: PMC11853595 DOI: 10.1093/oncolo/oyae343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/04/2024] [Indexed: 02/27/2025] Open
Abstract
Patients with advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) have impaired nutritional and physical performance due to the cancer pathophysiology and its treatment. The NUTRIGETNE study sought to characterize the nutritional status of patients with advanced GEP-NENs in Spain. This is a cross-sectional study that included patients with advanced GEP-NENs receiving active oncological treatment. Patients had a complete physical examination, anthropometry, bioelectrical impedance, dynamometry, laboratory analysis, and a comprehensive nutritional risk assessment. Malnutrition was defined according to Global Leadership Initiative on Malnutrition (GLIM) criteria. The study included 399 patients out of the 400 planned (Pearson's χ2; α 0.05). Median age was 62 years (22-83). Tumors most commonly originated in the small intestine (43.9%) and the pancreas (41.6%), 94.7% were metastatic, and 36.7%, 49.4%, and 12.5% were G1, G2, and G3, respectively. Malnutrition prevalence was 61.9% (25.8% moderate; 36.1% severe), mainly due to low muscle mass (50.9%), which was the most prevalent GLIM phenotypic criteria. Moreover, malnutrition showed a correlation with decreased hand grip strength (mean 23 vs 31.9 kg; P <.001) and phase angle (median 5o vs 5.6o; P <.001). The prevalence of sarcopenia was 15%. Malnutrition was more frequent in patients with diabetes (74.4% vs 56.7%; P <.001), NECs (82.1% vs 60.3%; P =.062), and in those treated with chemotherapy (71.2% vs 59.7%; P =.058), whereas it did not correlate with tumor origin (P =.507), histological grade (P =.781), or functionality (P =.465). Malnutrition was correlated to body mass index (BMI) (P =.015), although it was also diagnosed in a high proportion of patients with no weight loss (63%, 54.1%, and 65.1% of patients with normal BMI, overweight, and obesity, respectively). Cachexia was present in 109 (27.3%) patients. Malnutrition is very prevalent and commonly underdiagnosed in patients with GEP-NENs. It is associated with sarcopenia and a worse QoL, requiring a multifactorial nutritional assessment. Certain factors such as the presence of diabetes may require closer monitoring due to a higher risk of malnutrition.
Collapse
Affiliation(s)
- Maribel del Olmo-García
- Endocrinology and Nutrition Department, Endocrinology, Nutrition and Diet Therapy Research Unit, University and Polytechnic Hospital La Fe, Valencia, 46026, Spain
- Departamento de Medicina, Universidad de Valencia, Valencia, 46010, Spain
| | - Lorena Hernandez-Rienda
- Endocrinology and Nutrition Department, Endocrinology, Nutrition and Diet Therapy Research Unit, University and Polytechnic Hospital La Fe, Valencia, 46026, Spain
| | - Rocio Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Imas12, Facultad de Medicina, UCM, Madrid, 28041, Spain
| | - Jorge Hernando
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Ana Custodio
- Medical Oncology Department, Hospital Universitario La Paz, Instituto de Investigación Biomédica Hospital Universitario La Paz (IdiPAZ), Madrid, 28046, Spain
| | - Beatriz Anton-Pascual
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Imas12, Facultad de Medicina, UCM, Madrid, 28041, Spain
| | - Marta Gomez
- Nutritional Support Unit, Vall Hebron University Hospital, Barcelona, 08035, Spain
| | - Samara Palma Milla
- Nutrition and Endocrinology Service, Hospital Universitario La Paz, Madrid, 28046, Spain
| | - Lorena Suarez
- Department of Endocrinology and Nutrition, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, 33011, Spain
| | - Marta Bellver
- Clinical Nutrition Unit, Institut Català d’Oncologia (ICO), l’Hospitalet de Llobregat, Barcelona, 08908, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), l’Hospitalet de Llobregat, Barcelona, 08908, Spain
| | - Vicente Alonso
- Medical Oncology Department, Instituto Aragones de Investigacion Sanitaria, Hospital Universitario Miguel Servet, Zaragoza, 50009, Spain
| | - Raquel Serrano
- Medical Oncology Department, Hospital Universitario Reina Sofía, Instituto Maimonides de Investigación Biomedica (IMIBIC), Córdoba, 14004, Spain
| | - Nuria Valdés
- Endocrinology and Nutrition Department, University Hospital Cruces, Barakaldo, 48903, Spain
- Biobizkaia CIBERDEM/CIBERER, EndoERN. UPV/EHU, Barakaldo, 48903, Spain
| | - Marcos Melian
- Medical Oncology Department, Instituto Valenciano de Oncología (IVO), Valencia, 46009, Spain
| | - Beatriz Febrero
- Department of Endocrine Surgery, General Surgery Service, Virgen de la Arrixaca University Hospital, Murcia, 30120, Spain
| | | | - Josefina Biarnes
- Endocrinology and Nutrition, Hospital Universitari de Girona Dr. Josep Trueta, Girona, 17007, Spain
| | - José Ángel Díaz-Pérez
- Endocrinology and Nutrition, Hospital Universitario Clínico San Carlos, Madrid, 28040, Spain
| | | | - Carlos Lopez
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, Spain
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, IDIVAL, UNICAN, Santander, 39008, Spain
| | - Miguel Ángel Martínez Olmos
- Endocrinology and Nutrition Department, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, A Coruña, 15706, Spain
- Molecular Endocrinology Group-Health Research Institute of Santiago de Compostela-IDIS, Santiago de Compostela, A Coruña, 15706, Spain
- CIBER de Fisiopatología de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Francisco Merino-Torres
- Endocrinology and Nutrition Department, Endocrinology, Nutrition and Diet Therapy Research Unit, University and Polytechnic Hospital La Fe, Valencia, 46026, Spain
- Departamento de Medicina, Universidad de Valencia, Valencia, 46010, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Vall Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - María Argente Pla
- Endocrinology and Nutrition Department, Endocrinology, Nutrition and Diet Therapy Research Unit, University and Polytechnic Hospital La Fe, Valencia, 46026, Spain
| |
Collapse
|
32
|
Courtel T, Orbach D, Lacour B, Roumy M, Hescot S, Desandes E, Philippe-Chomette P, Sarnacki S, Irtan S, Dijoud F, Kubicek P, Brisse H, Fresneau B, Pire A, Réguerre Y, Mallebranche C. Childhood pancreatic neuroendocrine neoplasms: A national experience. Pediatr Blood Cancer 2025; 72:e31258. [PMID: 39135330 DOI: 10.1002/pbc.31258] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 12/24/2024]
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) diagnosed in childhood are very rare, with few data available. The aim was to describe the clinical presentation and behavior of children with pNENs at a national level. METHODS National multicenter retrospective study of all patients, aged from 0 to 17 years at diagnosis, treated from 2011 to 2020 for a pNEN and registered in the French National Registry of Childhood Cancers or FRACTURE database. RESULTS Fifteen patients, 13 well-differentiated pancreatic neuroendocrine tumors (pNETs) and two neuroendocrine carcinomas (pNECs), were selected. Median age at diagnosis was 14 years (range, 7-17). Eight patients, all with localized disease, had a cancer predisposition syndrome (CPS), including five cases diagnosed during systematic screening. Five (31%) had metastatic disease at diagnosis: three grade 2 pNETs and two pNECs. First line therapy included exclusive pancreatectomy (seven cases, all M0), active surveillance (three cases, all M0), medical therapies (somatostatin analogues, chemotherapy; four cases, all M1), and surgery with medical therapy (one M1 case). Three-year progression-free survival was 57% (confidence interval [CI] 95%: 27-78) and was significantly better for patients with low-grade well differentiated (73 vs. 0%; p < 10-4) and localized (76 vs. 20%; p = .02) tumors. The two patients with pNECs died. Three-year overall survival was 92% (CI95%: 59-99) and was significantly better in patients with low-grade tumor (100 vs. 50%; p = 10-4). CONCLUSION Childhood pNENs occur more frequently in adolescents with CPS. Localized low-grade pNETs in children have a very good prognosis, whereas the treatment of high-grade and metastatic pNETs/pNECs should be better defined.
Collapse
Affiliation(s)
- Tiphaine Courtel
- Unité d'Onco-Hémato-Immunologie pédiatrique, CHU Angers, Angers, France
| | - Daniel Orbach
- SIREDO Oncology Center (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Paris-Saclay University (PSL), Paris, France
| | - Brigitte Lacour
- French National Registry of Childhood Solid Tumors, CHU Nancy, Nancy, France
- Inserm UMR 1153, Centre of Research in Epidemiology and StatisticS (CRESS), Paris University, Epidemiology of Childhood and Adolescent Cancers Team (EPICEA), Paris, France
| | - Marianne Roumy
- Plateforme de recherche clinique pédiatrique, CHU Angers, Angers, France
| | - Ségolène Hescot
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
| | - Emmanuel Desandes
- French National Registry of Childhood Solid Tumors, CHU Nancy, Nancy, France
- Inserm UMR 1153, Centre of Research in Epidemiology and StatisticS (CRESS), Paris University, Epidemiology of Childhood and Adolescent Cancers Team (EPICEA), Paris, France
| | - Pascale Philippe-Chomette
- Department of Pediatric Surgery, Assistance Publique des Hôpitaux de Paris, Hôpital Robert Debré, Paris, France
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Urology and Transplantation, Necker-Enfants Malades Hospital, APHP, Université de Paris Cité, Paris, France
| | - Sabine Irtan
- APHP-Sorbonne University, Paris, France
- Department of Pediatric Visceral and Neonatal Surgery, Armand-Trousseau Children's Hospital, APHP, Paris, France
| | | | - Pierre Kubicek
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | - Hervé Brisse
- Department of Imaging, Institut Curie, PSL University, Paris, France
| | - Brice Fresneau
- Gustave Roussy, Université Paris-Saclay, Department of Children and Adolescent Oncology, Paris-Saclay University, Paris-Sud University, CESP, INSERM, Villejuif, France
| | - Aurore Pire
- Department of Pediatric Surgery, Urology and Transplantation, Necker-Enfants Malades Hospital, APHP, Université de Paris Cité, Paris, France
| | - Yves Réguerre
- Unité d'oncologie et d'hématologie pédiatrique, CHU Saint Denis de la Réunion, Bellepierre, France
| | - Coralie Mallebranche
- Unité d'Onco-Hémato-Immunologie pédiatrique, CHU Angers, Angers, France
- Université d'Angers, Université de Nantes, CHU Angers, INSERM, CRCI2NA, SFR ICAT, Angers, France
| |
Collapse
|
33
|
Nishida T. Subcutaneous Lanreotide Depot: A Diagnostic Pitfall in Neuroendocrine Tumor Imaging. Cureus 2025; 17:e79436. [PMID: 40130122 PMCID: PMC11931402 DOI: 10.7759/cureus.79436] [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: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Lanreotide depot, a long-acting somatostatin analog, improves progression-free survival in neuroendocrine tumor (NET) patients. However, persistent subcutaneous depots can appear as metastatic lesions on imaging, potentially leading to misdiagnosis. We report the case of a 70-year-old man with grade 2 NET (Ki-67: 5.9%) undergoing six years of lanreotide treatment. Follow-up imaging revealed stable primary disease. However, nodular lesions in the buttock were identified on the CT scan performed eight months after the start of treatment. A retrospective review of a CT scan taken two months after treatment initiation revealed that these nodules were already present and had gradually increased in number, allowing for their correct identification as persistent lanreotide depots. This case underscores the need for heightened diagnostic awareness to prevent unnecessary interventions and ensure accurate management.
Collapse
Affiliation(s)
- Tsutomu Nishida
- Department of Gastroenterology, Toyonaka Municipal Hospital, Toyonaka, JPN
| |
Collapse
|
34
|
Melhorn P, Raderer M, Kiesewetter B. Selecting systemic treatment for metastatic neuroendocrine tumors of the lung-current evidence and clinical implications. Cancer Treat Rev 2025; 133:102878. [PMID: 39787793 DOI: 10.1016/j.ctrv.2024.102878] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Neuroendocrine tumors (NET) of the lung are a slowly growing subtype of lung cancer that has a different treatment paradigm than aggressive and more common forms of lung neuroendocrine neoplasms (NEN) like small cell lung cancer (SCLC). Current guidelines for metastatic lung NET advocate a handful of treatment options, including somatostatin analogs (SSA), everolimus, temozolomide- or platin-based chemotherapy, and peptide receptor radionuclide therapy (PRRT). However, there is no clear treatment sequence, and the therapy of choice may depend on several factors such as tumor grade / growth rate, tumor burden / symptoms, disease progression status, and somatostatin receptor (SSTR) expression. In order to tailor treatment to each individual patient, the latest scientific findings and patient-specific clinical features must be considered together. This review critically evaluates the available evidence with regards to relevant patient characteristics, inclusion and exclusion criteria, and outcome metrics of clinical trials given the presumed natural disease course. Specific patient subgroups with an unmet therapeutic need are identified and discussed in the context of ongoing clinical trials.
Collapse
Affiliation(s)
- Philipp Melhorn
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Markus Raderer
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Barbara Kiesewetter
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria.
| |
Collapse
|
35
|
Lauricella E, Vilisova S, Chaoul N, Giglio A, D'Angelo G, Porta C, Cives M. The current status of somatostatin analogs in the treatment of neuroendocrine tumors and future perspectives. Expert Rev Neurother 2025; 25:245-258. [PMID: 39415322 DOI: 10.1080/14737175.2024.2417419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Somatostatin analogs (SSAs) were developed as antisecretory agents to palliate hormonal symptoms in patients with functioning neuroendocrine tumors (NETs). Their antiproliferative activity has been established in the phase 3 PROMID and CLARINET trials. SSAs currently represent the standard first-line therapy for the majority of well-differentiated G1/G2 gastroenteropancreatic NETs as well as for pulmonary NETs. AREAS COVERED An update on the clinical applications of established SSAs for the treatment of NETs is provided. Perspectives on emerging nonpeptide SSAs such as paltusotine and innovative formulations of octreotide (CAM2029) are included. EXPERT OPINION SSAs represent the cornerstone of treatment for both functioning and nonfunctioning NETs. While standard-dose SSAs have a defined place in the therapeutic algorithm of well-differentiated NETs, uncertainties remain on how to best integrate above-label doses of SSAs in the treatment sequence, particularly when tumor control is the goal. Octreotide and lanreotide appear to be clinically interchangeable, and no signs of superiority of one agent over the other has been observed so far. Whether SSAs may be exploited in the maintenance setting following more aggressive treatments, whether continuing SSAs beyond-progression after first-line therapy could be an effective treatment strategy, and whether new-generation SSAs such as pasireotide could overcome resistance to established SSAs are key areas of investigation.
Collapse
Affiliation(s)
- Eleonora Lauricella
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Sofija Vilisova
- Department of Oncology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Nada Chaoul
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Andrea Giglio
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Gabriella D'Angelo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| |
Collapse
|
36
|
Fujimori N, Fujita N, Murakami M, Ohno A, Matsumoto K, Teramatsu K, Ueda K, Wada N, Takao S, Okamoto D, Ishigami K, Ito T, Ogawa Y. Usefulness of Semiautomated 3D Volumetric Assessment of Liver Tumor Burden for Patients With Unresectable Pancreatic Neuroendocrine Tumor: A Pilot Study. Pancreas 2025; 54:e122-e129. [PMID: 39928889 DOI: 10.1097/mpa.0000000000002413] [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] [Indexed: 02/12/2025]
Abstract
OBJECTIVES In patients with metastatic pancreatic neuroendocrine tumors (PanNETs), the Ki-67 index is objectively assessed by pathologists; however, liver tumor burden (LTB) depends on the subjective judgment of physicians. This study aimed to elucidate the usefulness of the semi-automated 3D volumetric assessment of LTB in patients with PanNET. MATERIALS AND METHODS We retrospectively reviewed 29 patients (40 computed tomographies [CTs]) with metastatic PanNETs. LTB was measured using a semiautomated 3D volumetric software program (volumetric assessment) or evaluated independently by 6 clinicians using CT imaging (visual assessment). The treatment map was classified into 3 groups based on LTB and Ki-67 index. RESULTS Visual and volumetric assessments of the LTB were well correlated. The LTB was significantly higher on visual assessment than volumetric assessment (P < 0.01). Categorization on the map was consistent between the visual and volumetric evaluations in 23 patients (equal group). The remaining 6 patients were overestimated by visual assessment (overestimated group). Progression-free survival was significantly longer in patients in the 'equal group' than the 'overestimated group' (981 vs 366 days, P < 0.01). CONCLUSIONS This pilot study revealed a good correlation between visual and volumetric assessments, and visual assessment overestimated LTB, compared to volumetric assessment.
Collapse
Affiliation(s)
- Nao Fujimori
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhiro Fujita
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Murakami
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihisa Ohno
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhide Matsumoto
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhito Teramatsu
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keijiro Ueda
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriaki Wada
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiichiro Takao
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Okamoto
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kousei Ishigami
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Yoshihiro Ogawa
- From the Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
37
|
Faggiano A, Russo F, Zamponi V, Sesti F, Puliani G, Modica R, Malandrino P, Ferraù F, Rinzivillo M, Di Muzio M, Di Simone E, Panattoni N, Dolce P, Lauretta R, Di Iasi G, Prinzi A, Alessi Y, Feola T, Mazzilli R, Appetecchia M, Giannetta E, Panzuto F, Colao A. Impact of dyslipidemia and lipid-lowering therapy with statins in patients with neuroendocrine tumors. J Neuroendocrinol 2025; 37:e13485. [PMID: 39726194 PMCID: PMC11791004 DOI: 10.1111/jne.13485] [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: 09/07/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Dyslipidemia is a potential unfavorable prognostic factor in neuroendocrine tumors (NETs); conversely, statins proved to have antiproliferative effects in NET cell lines and could be a helpful therapeutic strategy for these patients. The main objective of this observational cohort retrospective study is to explore the associations between dyslipidemia and NET progression and evaluate the potential influence of statins in this context. 393 patients with histologically confirmed gastroenteropancreatic or bronchopulmonary NETs from six Italian centres didicated to NET diagnosis and therapy were included. The cohort included 123 patients with dyslipidemia, 81 of which were taking statins. Clinicopathological data, including patient demographics, tumor characteristics, and treatment details as well as the prevalence, timing of dyslipidemia and hypolipemic therapy were collected. The main outcome measure used is progression-free survival (PFS). Among the 393 patients, 123 (31.3%) had dyslipidemia. Statins were used by 81 (65.8%) dyslipidemic patients, mostly atorvastatin. Median PFS was 87 months overall, 124 months in non-dyslipidemic patients, and 72 months in dyslipidemic patients (p = .268). Dyslipidemic patients on statins had a significantly better median PFS (108 months) than those not on statins (26 months; p = .024). Recurrence-free survival (RFS) was also evaluated, but no significant differences were found. In conclusion, while PFS was lower in dyslipidemic patients compared to non-dyslipidemic patients, the difference was not statistically significant. Statin therapy was associated with improved PFS among dyslipidemic patients, suggesting a potential antiproliferative effect of statins in NETs. These findings warrant further investigation to substantiate the role of statins in the management of NETs.
Collapse
Affiliation(s)
- Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | - Flaminia Russo
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | - Virginia Zamponi
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | - Franz Sesti
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Giulia Puliani
- Oncological Endocrinology UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Roberta Modica
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and SurgeryFederico II University of NaplesNaplesItaly
| | - Pasqualino Malandrino
- Endocrinology Unit, Department of Clinical and Experimental MedicineGaribaldi ‐ Nesima Medical Center, University of CataniaCataniaItaly
| | - Francesco Ferraù
- Department of Human Pathology of Adulthood and Childhood ‘G. Barresi’University of MessinaMessinaItaly
| | - Maria Rinzivillo
- Digestive Disease Unit, Department of Medical‐Surgical Sciences and Translational Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | - Marco Di Muzio
- Department of Clinical and Molecular MedicineSapienza University of RomeRomeItaly
| | - Emanuele Di Simone
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Nicolò Panattoni
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Pasquale Dolce
- Department of Translational Medical ScienceFederico II UniversityNaplesItaly
| | - Rosa Lauretta
- Oncological Endocrinology UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gianfranco Di Iasi
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and SurgeryFederico II University of NaplesNaplesItaly
| | - Antonio Prinzi
- Endocrinology Unit, Department of Clinical and Experimental MedicineGaribaldi ‐ Nesima Medical Center, University of CataniaCataniaItaly
| | - Ylenia Alessi
- Department of Biomedical, Dental and Morphological and Functional Imaging SciencesUniversity of MessinaMessinaItaly
| | - Tiziana Feola
- Department of Experimental MedicineSapienza University of RomeRomeItaly
- NeuroendocrinologyNeuromed Institute, IRCCSPozzilliItaly
| | - Rossella Mazzilli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | | | - Elisa Giannetta
- Department of Experimental MedicineSapienza University of RomeRomeItaly
| | - Francesco Panzuto
- Digestive Disease Unit, Department of Medical‐Surgical Sciences and Translational Medicine, European Neuroendocrine Tumor Society (ENETS) Center of ExcellenceSant'Andrea University Hospital, Sapienza University of RomeRomeItaly
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and SurgeryFederico II University of NaplesNaplesItaly
- UNESCO Chair “Education for Health and Sustainable Development”Federico II UniversityNaplesItaly
| |
Collapse
|
38
|
Tran CG, Sherman SK, Chandrasekharan C, Howe JR. Surgical Management of Neuroendocrine Tumor Liver Metastases. Hematol Oncol Clin North Am 2025; 39:37-53. [PMID: 39510676 DOI: 10.1016/j.hoc.2024.08.001] [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] [Indexed: 11/15/2024]
Abstract
Patients with neuroendocrine tumor liver metastases (NETLMs) may develop carcinoid syndrome, carcinoid heart disease, or other symptoms from overproduction of hormones. Hepatic resection and cytoreduction is the most direct treatment of NETLMs in eligible patients, and cytoreduction improves symptoms, may reduce the sequelae of carcinoid syndrome, and extends survival. Parenchymal-sparing procedures, such as ablation and enucleation, should be considered during cytoreduction to maximize treatment of multifocal tumors while preserving healthy liver tissue. For patients with large hepatic tumor burdens, high-grade disease, or comorbidities precluding surgery, liver-directed and systemic therapies can be used to palliate symptoms and improve progression-free survival.
Collapse
Affiliation(s)
- Catherine G Tran
- Department of Surgery, Division of Colorectal Surgery, University of Minnesota, Minneapolis, MN
| | - Scott K Sherman
- Department of Surgery, Division of Surgical Oncology and Endocrine Surgery, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | | | - James R Howe
- Department of Surgery, Division of Surgical Oncology and Endocrine Surgery, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| |
Collapse
|
39
|
Tacelli M, Partelli S, Falconi M, Arcidiacono PG, Capurso G. Pancreatic Neuroendocrine Neoplasms: Classification and Novel Role of Endoscopic Ultrasound in Diagnosis and Treatment Personalization. United European Gastroenterol J 2025; 13:34-43. [PMID: 39540703 PMCID: PMC11866312 DOI: 10.1002/ueg2.12710] [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: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
The incidence and prevalence of pancreatic neuroendocrine neoplasms are steadily increasing. These tumors are highly heterogeneous, with treatment options ranging from observation to surgery, and various medical therapies. The choice of treatment is influenced by factors such as tumor stage, grade (proliferative activity), and the presence of hormone-related syndromes. Endoscopic ultrasound (EUS) is becoming increasingly valuable for assessing pancreatic neuroendocrine neoplasms, offering detailed morphological, vascular, and functional information through techniques such as contrast enhancement and elastography. It also allows biopsies that are useful for both histopathological and molecular analyses. These tumors are highly heterogeneous, with treatment options ranging from observation to various medical therapies and surgery. Recent data suggest that small, non-functioning PanNENs with low proliferation rates may be safely monitored, whereas more aggressive or functioning tumors typically require surgery. EUS-guided ablation is a promising alternative for patients with functional pancreatic neuroendocrine neoplasms who are unsuitable for surgery, although randomized trials are needed. In non-resectable pancreatic neuroendocrine neoplasms, treatment options include somatostatin analogs, targeted therapies (e.g., everolimus, sunitinib), chemotherapy, and radioligand therapy. This review discusses key factors in planning personalized treatment strategies for pancreatic neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato‐Biliary Endoscopy and Endosonography DivisionPancreas Translational and Clinical Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Stefano Partelli
- Pancreatic Surgery UnitPancreas Translational and Clinical Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
- “Vita‐Salute” San Raffaele UniversityMilanItaly
| | - Massimo Falconi
- Pancreatic Surgery UnitPancreas Translational and Clinical Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
- “Vita‐Salute” San Raffaele UniversityMilanItaly
| | - Paolo Giorgio Arcidiacono
- Pancreato‐Biliary Endoscopy and Endosonography DivisionPancreas Translational and Clinical Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
- “Vita‐Salute” San Raffaele UniversityMilanItaly
| | - Gabriele Capurso
- Pancreato‐Biliary Endoscopy and Endosonography DivisionPancreas Translational and Clinical Research CenterIRCCS San Raffaele Scientific InstituteMilanItaly
- “Vita‐Salute” San Raffaele UniversityMilanItaly
| |
Collapse
|
40
|
McBrien C, O’Connell DJ. The Use of Biologics for Targeting GPCRs in Metastatic Cancers. BIOTECH 2025; 14:7. [PMID: 39982274 PMCID: PMC11843943 DOI: 10.3390/biotech14010007] [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: 12/18/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
A comprehensive review of studies describing the role of G-protein coupled receptor (GPCR) behaviour contributing to metastasis in cancer, and the developments of biotherapeutic drugs towards targeting them, provides a valuable resource toward improving our understanding of the opportunities to effectively target this malignant tumour cell adaptation. Focusing on the five most common metastatic cancers of lung, breast, colorectal, melanoma, and prostate cancer, we highlight well-studied and characterised GPCRs and some less studied receptors that are also implicated in the development of metastatic cancers. Of the approximately 390 GPCRs relevant to therapeutic targeting, as many as 125 of these have been identified to play a role in promoting metastatic disease in these cancer types. GPCR signalling through the well-characterised pathways of chemokine receptors, to emerging data on signalling by orphan receptors, is integral to many aspects of the metastatic phenotype. Despite having detailed information on many receptors and their ligands, there are only thirteen approved therapeutics specifically for metastatic cancer, of which three are small molecules with the remainder including synthetic and non-synthetic peptides or monoclonal antibodies. This review will cover the existing and potential use of monoclonal antibodies, proteins and peptides, and nanobodies in targeting GPCRs for metastatic cancer therapy.
Collapse
Affiliation(s)
| | - David J. O’Connell
- School of Biomolecular & Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| |
Collapse
|
41
|
Halperin R, Tirosh A. Progress report on multiple endocrine neoplasia type 1. Fam Cancer 2025; 24:15. [PMID: 39826015 PMCID: PMC11742904 DOI: 10.1007/s10689-025-00440-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) syndrome is an autosomal dominant disorder caused by a germline pathogenic variant in the MEN1 tumor suppressor gene. Patients with MEN1 have a high risk for primary hyperparathyroidism (PHPT) with a penetrance of nearly 100%, pituitary adenomas (PitAd) in 40% of patients, and neuroendocrine neoplasms (NEN) of the pancreas (40% of patients), duodenum, lung, and thymus. Increased MEN1-related mortality is mainly related to duodenal-pancreatic and thymic NEN. Management of PHPT differs from that of patients with sporadic disease, as the surgical approach in MEN1-related PHPT includes near-total or total parathyroidectomy because of multigland hyperplasia in most patients and the consequent high risk of recurrence. NEN management also differs from patients with sporadic disease due to multiple synchronous and metasynchronous neoplasms. In addition, the lifelong risk of developing NEN requires special considerations to avoid excessive surgeries and to minimize damage to the patient's function and well-being. This progress report will outline current insights into surveillance and management of the major clinical manifestation of MEN1 syndrome in children and adults with MEN1 diagnosis. In addition, we will discuss MEN1-like clinical presentation with negative MEN1-genetic workup and future clinical and research directions.
Collapse
Affiliation(s)
- Reut Halperin
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuroendocrine Oncology Unit, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan, Israel
- The Chaim Sheba Medical Center, ENTIRE - Endocrine Neoplasia Translational Research Center, Tel Aviv University Faculty of Medicine, 2 Sheba Road, Tel HaShomer, Ramat Gan, Israel
| | - Amit Tirosh
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Neuroendocrine Oncology Unit, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Ramat Gan, Israel.
- The Chaim Sheba Medical Center, ENTIRE - Endocrine Neoplasia Translational Research Center, Tel Aviv University Faculty of Medicine, 2 Sheba Road, Tel HaShomer, Ramat Gan, Israel.
| |
Collapse
|
42
|
Polici M, Caruso D, Masci B, Marasco M, Valanzuolo D, Dell'Unto E, Zerunian M, Campana D, De Santis D, Lamberti G, Iannicelli E, Prosperi D, Annibale B, Laghi A, Panzuto F, Rinzivillo M. Radiomics in advanced gastroenteropancreatic neuroendocrine neoplasms: Identifying responders to somatostatin analogs. J Neuroendocrinol 2025; 37:e13472. [PMID: 39564809 PMCID: PMC11750307 DOI: 10.1111/jne.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/15/2024] [Accepted: 11/02/2024] [Indexed: 11/21/2024]
Abstract
To evaluate a radiomic strategy for predicting progression in advanced gastroenteropancreatic neuroendocrine tumor (GEP-NET) patients treated with somatostatin analogs (SSAs). Fifty-eight patients with GEP-NETs and liver metastases, with baseline computerized tomography (CT) scans from June 2013 to November 2020, were studied retrospectively. Data collected included progression-free survival (PFS), overall survival (OS), tumor grading, death, and Ki67 index. Patients were categorized into progressive and non-progressive groups. Two radiologists performed 3D liver segmentation on baseline CT scans using 3DSlicer v4.10.2. One hundred six radiomic features were extracted and analyzed (T-test or Mann-Whitney). Radiomic feature efficacy was evaluated via receiver operating characteristic curves, and both univariate and multivariate logistic regression were used to develop predictive models. A significance level of p < .05 was maintained. Of 55 patients, 38 were progressive (median PFS and OS: 14 and 34 months, respectively), and 17 were non-progressive (median PFS and OS: 58 months each). Six radiomic features significantly differed between groups (p < .05), with an area under the curve (AUC) range of 0.64-0.74. Ki67 was the only clinical parameter significantly associated with progression risk (odds ratio (OR) = 1.14, p < .05). The combined radiomic features and Ki67 model proved most effective, showing an AUC of 0.814 (p = .008). The radiomic model alone did not reach statistical significance (p = .07). A combined model incorporating radiomic features and the Ki67 index effectively predicts disease progression in GEP-NET patients eligible for SSA treatment.
Collapse
Affiliation(s)
- Michela Polici
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Damiano Caruso
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Benedetta Masci
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Matteo Marasco
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Digestive Disease Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Daniela Valanzuolo
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Elisabetta Dell'Unto
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Digestive Disease Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Marta Zerunian
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Davide Campana
- Department of Medical and Surgical Sciences (DIMEC), Medical Oncology UnitAlma Mater Studiorum—University of Bologna, IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Domenico De Santis
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Medical Oncology UnitAlma Mater Studiorum—University of Bologna, IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Elsa Iannicelli
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Daniela Prosperi
- ENETS Center of Excellence, Nuclear Medicine Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Bruno Annibale
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Digestive Disease Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Andrea Laghi
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Radiology Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Francesco Panzuto
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Digestive Disease Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| | - Maria Rinzivillo
- Department of Medical‐Surgical Sciences and Translational Medicine, ENETS Center of Excellence, Digestive Disease Unit, Sant'Andrea University HospitalSapienza University of RomeRomeItaly
| |
Collapse
|
43
|
Ozsoy MS, Erol CI, Aydemir MA, Baysal H, Buyuker F, Seneldir H, Ekinci O, Eren T, Alimoglu O. Gastroenteropancreatic Neuroendocrine Tumors: Does Tumor Location Affect Prognosis? ARCHIVES OF IRANIAN MEDICINE 2025; 28:29-39. [PMID: 40001327 PMCID: PMC11862391 DOI: 10.34172/aim.33366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 11/11/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare entities. Generally, they can be localized anywhere in the gastrointestinal or hepatobiliary tract. The purpose of our study is to evaluate the effect of tumor location on prognosis in patients with GEP-NET undergoing surgery. Our secondary objective is to examine other factors affecting the prognosis of patients with GEP-NET. METHODS We retrospectively analyzed data from 30 patients with GEP-NET who underwent surgery in the General Surgery Clinic between 2012 and 2022. The gNET group (n=18) included tumors located in the gastrointestinal tract, while the pNET group (n=12) included tumors located in the hepatopancreatobiliary system. Surgical, laboratory, radiological, and pathological findings of the patients, as well as follow-up outcomes were recorded and statistically analyzed. RESULTS In subgroup comparison, tumor size was found to be larger in the pNET group (P=0.002). The statistical analysis of recurrence (16.7% versus 33.3%) and mortality rates (16.7% versus 41.7%) between the subgroups (P=0.329 and P=0.210, respectively) did not reveal a significant difference. When all patients were evaluated, it was observed that advanced age, presence of carcinoma diagnosis, higher tumor grade, advanced TNM stage, larger tumor size, presence of lymphovascular or perineural invasion, elevated mitotic index, higher Ki-67 index, and having received adjuvant therapy increased the rates of recurrence and mortality. CONCLUSION There was no statistically significant difference in survival outcomes between the GEP-NET groups located in the gastrointestinal tract and the hepatopancreatobiliary system.
Collapse
Affiliation(s)
- Mehmet Sait Ozsoy
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Cem Ilgin Erol
- Department of General Surgery, Erzurum City Hospital, Erzurum, Turkey
| | - Muhammet Ali Aydemir
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Hakan Baysal
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Fatih Buyuker
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Hatice Seneldir
- Department of Pathology, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Ozgur Ekinci
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Tunc Eren
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| | - Orhan Alimoglu
- Department of General Surgery, Faculty of Medicine, Istanbul Medeniyet University, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkey
| |
Collapse
|
44
|
Kasai Y, Ito T, Masui T, Nagai K, Anazawa T, Uchida Y, Ishii T, Umeshita K, Eguchi S, Soejima Y, Ohdan H, Hatano E. Liver transplantation for gastroenteropancreatic neuroendocrine liver metastasis: optimal patient selection and perioperative management in the era of multimodal treatments. J Gastroenterol 2025; 60:1-9. [PMID: 39547997 PMCID: PMC11717855 DOI: 10.1007/s00535-024-02166-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
Gastroenteropancreatic neuroendocrine tumors (NET) often metastasize to the liver. Although curative liver resection provides a favorable prognosis for patients with neuroendocrine liver metastasis (NELM), with a 5-year survival rate of 70-80%, recurrence is almost inevitable, mainly in the remnant liver. In Western countries, liver transplantation (LT) has been performed in patients with NELM, with the objective of complete removal of macro- and micro-NELMs. However, prognosis had been unsatisfactory, with 5-year overall survival and recurrence-free survival rates of approximately 50 and 30%, respectively. In 2007, the Milan criteria were proposed as indications for LT for NELM. The criteria included: (1) confirmed histology of NET-G1 or G2; (2) a primary tumor drained by the portal system and all extrahepatic diseases removed with curative resection before LT; (3) liver involvement ≤50%; (4) good response or stable disease for at least 6 months before LT; (5) age ≤ 55 years. A subsequent report demonstrated outstanding LT outcomes for NELM within the Milan criteria, with 5-year overall survival and recurrence rates of 97 and 13%, respectively. In Japan, living donor LT (LDLT) for NELM has been performed sporadically in only 16 patients by 2021 in Japan; however, no consensus has been reached on the indications or perioperative management of LDLT. This article presents the outcomes of these 16 patients who underwent LDLT in Japan and reviews the literature to clarify optimal indications and perioperative management of LDLT for NELM in the era of novel multimodal treatments.
Collapse
Affiliation(s)
- Yosuke Kasai
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takashi Ito
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Surgery, Kurashiki Central Hospital, Okayama, Japan
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoichiro Uchida
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takamichi Ishii
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koji Umeshita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
- Osaka International Cancer Institute, Osaka, Japan
| | - Susumu Eguchi
- Department of Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuji Soejima
- Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
45
|
Tobias J, Abou Azar S, Gujarathi R, Nordgren R, Vaghaiwalla T, Millis JM, Feinberg N, Liao CY, Keutgen XM. Surgery enhances the effectiveness of peptide receptor radionuclide therapy in metastatic gastroenteropancreatic neuroendocrine tumors. Surgery 2025; 177:108834. [PMID: 39395860 DOI: 10.1016/j.surg.2024.06.065] [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: 02/14/2024] [Revised: 05/11/2024] [Accepted: 06/02/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND With the advent of peptide receptor radionuclide therapy, the timing and sequence of surgery in the treatment of metastatic gastroenteropancreatic neuroendocrine tumors merits further study. We hypothesized that surgery before peptide receptor radionuclide therapy might enhance its effectiveness in patients with metastatic gastroenteropancreatic neuroendocrine tumors. METHODS Eighty-nine patients with metastatic well-differentiated gastroenteropancreatic neuroendocrine tumors treated with 177Lutetium-dotatate peptide receptor radionuclide therapy between 2018 and 2023 were included. Fifty-six patients underwent surgery (primary tumor resection and/or liver debulking) before peptide receptor radionuclide therapy and 33 patients did not. Primary outcome was progression-free survival according to Response Evaluation Criteria in Solid Tumors. Pretreatment dotatate positron emission tomography/computed tomography was used to calculate tumor volumes. RESULTS The surgery and no-surgery groups were well-matched. Median progression-free survival after peptide receptor radionuclide therapy was 15.6 months (interquartile range, 9.1-22.7 months) in the no-surgery group compared with 26.1 months (interquartile range, 12.7-38.1 months) in the surgery group (P = .04). On subgroup analysis, median progression-free survival was 18.1 months (interquartile range, 11.9-38.4 months) in patients who underwent primary tumor resection only compared with 26.2 months (interquartile range, 14.0-38.1 months) in patients who underwent liver debulking (P = .04). Tumor volume was lowest in patients who underwent liver debulking (median 146.07 mL3) compared with no surgery (median 626.42 mL3) (P = .001). On univariable analysis, a tumor volume <138.8 mL3 was associated with longer progression-free survival (hazard ratio, 2.03; 95% confidence interval, 0.95-4.34, P = .05), with a median progression-free survival of 38.1 months (interquartile range, 16.9-41.3 months) compared with 17.8 months (interquartile range, 10.8-28.7 months). CONCLUSION Surgery may enhance the effectiveness of 177Lutetium-dotatate in the treatment of metastatic well-differentiated gastroenteropancreatic neuroendocrine tumors. This positive effect may be the result of a lower tumor volume in patients after surgery. Our findings fortify the concept of using surgical debulking to improve systemic therapies such as peptide receptor radionuclide therapy.
Collapse
Affiliation(s)
- Joseph Tobias
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, Chicago, IL.
| | - Sara Abou Azar
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, Chicago, IL
| | - Rushabh Gujarathi
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - Rachel Nordgren
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | - Tanaz Vaghaiwalla
- Division of Endocrine Surgery, University of Miami Miller School of Medicine, Miami, FL
| | - J Michael Millis
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, Chicago, IL
| | - Nicholas Feinberg
- Section of Nuclear Medicine, Department of Radiology, University of Chicago, Chicago, IL
| | - Chih-Yi Liao
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | - Xavier M Keutgen
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, Chicago, IL
| |
Collapse
|
46
|
Zhao L, Cheng X, Zhao H, Zhao H, Di W, Mei Z. Long‑term survival after comprehensive treatment in a patient with advanced neuroendocrine neoplasm of the pancreas: A case report. Oncol Lett 2025; 29:49. [PMID: 39564371 PMCID: PMC11574579 DOI: 10.3892/ol.2024.14795] [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: 07/14/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
Neuroendocrine neoplasms of the pancreas (pNENs) are rare. In February 2021, a 54-year-old woman was diagnosed with pNEN and multiple metastases within the liver. The patient, diagnosed with grade G2 neuroendocrine neoplasm (T4N0M1), underwent an ultrasonography-guided liver biopsy and radiofrequency ablation. After receiving Sandostatin LAR in April 2021, side effects led to its discontinuation after seven cycles. Following two sessions of radiofrequency ablation, the patient's condition was stable. However, disease progression was noted in September 2023, resulting in hemodialysis and closed peritoneal drainage. Surufatinib was administered, stabilizing the tumor by November 2023. The patient underwent transarterial chemoembolization due to a large tumor burden, with subsequent MRCP showing stability from diagnosis in February 2021 to June 2024. The present case report highlights the role of tailored treatment strategies considering patient comorbidities and tumor biology, and the significance of secondary puncture biopsy, which, despite not being pursued by the patient in the present study due to the associated risks, may provide survival benefits for patients with advanced or metastatic pNEN.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xin Cheng
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Hongbin Zhao
- Department of General Surgery, Huaihai Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Haifei Zhao
- Department of Imaging, Huaihai Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Wenyu Di
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| | - Zhihong Mei
- Department of Radiotherapy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, P.R. China
| |
Collapse
|
47
|
Howe JR, Menda Y, Chandrasekharan C, Bellizzi AM, Quelle DE, O'Dorisio MS, Dillon JS. The University of Iowa Neuroendocrine Tumor Clinic. Endocr Pract 2025; 31:4-18. [PMID: 39349242 PMCID: PMC11700786 DOI: 10.1016/j.eprac.2024.09.018] [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: 04/02/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
The Iowa Neuroendocrine Tumor (NET) Clinic was founded and developed by two remarkable physicians, Thomas and Sue O'Dorisio. Tom was an Endocrinologist and close friend and colleague of Aaron Vinik. Both men were pioneers in studies of gastrointestinal hormones and the management of patients with NETs. Sue was a Pediatric Oncologist and research scientist with great expertise in new drug development and clinical trials. She and Tom were leaders in bringing somatostatin analogs and somatostatin-conjugated radioligands to the clinic for the therapy and diagnosis of NETs. All three physicians received lifetime achievement awards for their contributions to the field of NETs. This is the story of how the Iowa NET Clinic developed over the years to become a model for the multidisciplinary mantagement of patients with NETs, culminating in its designation as a European Neuroendocrine Tumor Society NET Center of Excellence, and the receipt of a Specialized Project of Research Excellence (SPORE) grant for the study of NETs from the National Institutes of Health.
Collapse
Affiliation(s)
- James R Howe
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| | - Yusuf Menda
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | | - Andrew M Bellizzi
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Dawn E Quelle
- Departments of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - M Sue O'Dorisio
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Joseph S Dillon
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
48
|
Meng QH, Halfdanarson TR, Bornhorst JA, Jann H, Shaheen S, Shi RZ, Schwabe A, Stade K, Halperin DM. Circulating Chromogranin A as a Surveillance Biomarker in Patients with Carcinoids-The CASPAR Study. Clin Cancer Res 2024; 30:5559-5567. [PMID: 39453770 DOI: 10.1158/1078-0432.ccr-24-1875] [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: 06/17/2024] [Revised: 08/09/2024] [Accepted: 10/23/2024] [Indexed: 10/27/2024]
Abstract
PURPOSE Gastroenteropancreatic neuroendocrine tumors (GEP-NET) are relatively indolent but can be more aggressive. The current recommendations for using serum chromogranin A (CgA) for patients with GEP-NET are equivocal. This study was designed to validate an automated CgA immunofluorescence assay for monitoring disease progression in patients with GEP-NET. PATIENTS AND METHODS A prospective, multicenter, blinded observational study was designed to validate an automated CgA immunofluorescence assay for monitoring disease progression in patients with GEP-NET. Tumor progression was evaluated with RECIST 1.1 by CT/MRI. An increase ≥50% above the prior CgA concentration to a value >100 ng/mL in the following CgA concentration was considered positive. RESULTS A total of 153 patients with GEP-NET were enrolled. Using the prespecified cut-off of CgA change for tumor progression, specificity was 93.4% (95% confidence interval, 90.4%-95.5%; P < 0.001), sensitivity 34.4% (25.6%-44.3%), positive predictive value 57.9% (45.0-69.8), negative predictive value 84.3% (80.5-87.6), and AUC 0.73 (0.67-0.79). CONCLUSIONS Changes in serial measurements of serum CgA had a favorable specificity and negative predictive value, making this test a useful adjunct to routine radiographic monitoring.
Collapse
Affiliation(s)
- Qing H Meng
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Joshua A Bornhorst
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Henning Jann
- Division of Hepatology and Gastroenterology, Medical Department, Charité-Universitätsmedizin, Berlin, Germany
| | - Shagufta Shaheen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Run Zhang Shi
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Andrej Schwabe
- B·R·A·H·M·S GmbH, part of Thermo Fisher Scientific, Hennigsdorf, Germany
| | - Katrin Stade
- B·R·A·H·M·S GmbH, part of Thermo Fisher Scientific, Hennigsdorf, Germany
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
49
|
Izumo W, Higuchi R, Furukawa T, Shiihara M, Uemura S, Yazawa T, Yamamoto M, Honda G. The importance of microvascular invasion in patients with non-functioning pancreatic neuroendocrine neoplasm. Langenbecks Arch Surg 2024; 410:8. [PMID: 39676094 DOI: 10.1007/s00423-024-03563-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND/OBJECTIVES The oncological importance of lymphatic, microvascular, and perineural invasions and their association with outcomes in patients with non-functioning pancreatic neuroendocrine neoplasm (NF-PanNEN) remains unclear. We aimed to investigate the role of these factors in the prognosis of patients with NF-PanNEN. METHODS We retrospectively analyzed 115 patients who underwent curative resection and were pathologically and clinically diagnosed with NF-PanNEN. We evaluated the relationship between clinicopathological factors and recurrence. RESULTS Thirty (26%), 38 (33%), and 11 (10%) patients had lymphatic, microvascular, and nerve invasions, respectively. Twenty-one patients (18%) experienced recurrence, with a median time to recurrence of 2.6 years (range: 0.3-8.2). The 3-, 5-, and 10-year recurrence-free survival (RFS) rates were 88.3%, 84.4%, and 79.1%, respectively. In multivariate analyses, World Health Organization Grade G2-3 (vs. G1, hazard ratio (HR): 16.2), T factor T3-4 (vs. T1-2, HR: 5.2), and the presence of microvascular invasion (vs. absence, HR: 5.6) were significant risk factors for RFS. When these risk factors were assigned as risk score of three, one, and one points depending on the HR, the 5-year recurrence rates in patients with risk score groups 0-1 and 2-5 were 98.6% and 53.3%, (P < 0.001). Moreover, only the presence of microvascular invasion significantly increased the likelihood of recurrence within 3 years. CONCLUSIONS The presence of microvascular invasion is an independent risk factor for recurrence in patients with NF-PanNEN. Our risk scoring system, which includes "the presence of microvascular invasion," may be useful for predicting recurrence.
Collapse
Affiliation(s)
- Wataru Izumo
- Department of Surgery, Institute of Gastroenterology, Tokyo Womens Medical University, 8-1 Kawada-cho, Tokyo, Shinjuku City, Japan
| | - Ryota Higuchi
- Department of Surgery, Institute of Gastroenterology, Tokyo Womens Medical University, 8-1 Kawada-cho, Tokyo, Shinjuku City, Japan.
- Division of Gastroenterological Surgery, Tokyo Womens Medical University Yachiyo Medical Center, Yachiyo City, Chiba, Japan.
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Shiihara
- Department of Surgery, Institute of Gastroenterology, Tokyo Womens Medical University, 8-1 Kawada-cho, Tokyo, Shinjuku City, Japan
| | - Shuichiro Uemura
- Department of Surgery, Institute of Gastroenterology, Tokyo Womens Medical University, 8-1 Kawada-cho, Tokyo, Shinjuku City, Japan
| | - Takehisa Yazawa
- Gastrointestinal surgery, Utsunomiya Memorial Hospital, Utsunomiya, Japan
| | - Masakazu Yamamoto
- Gastrointestinal surgery, Utsunomiya Memorial Hospital, Utsunomiya, Japan
| | - Goro Honda
- Department of Surgery, Institute of Gastroenterology, Tokyo Womens Medical University, 8-1 Kawada-cho, Tokyo, Shinjuku City, Japan
| |
Collapse
|
50
|
Cruz-Diaz WE, Paitan V, Medina J, Flores R, Haro-Varas J, Mantilla R, Castro-Oliden V. Temozolomide and capecitabine regimen as first-line treatment in advanced gastroenteropancreatic neuroendocrine tumors at a Latin American reference center. World J Gastrointest Oncol 2024; 16:4675-4684. [PMID: 39678797 PMCID: PMC11577364 DOI: 10.4251/wjgo.v16.i12.4675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND Numerous studies have indicated that the temozolomide and capecitabine regimen (TEMCAP) exhibits a certain level of efficacy in treating advanced, well-differentiated gastroenteropancreatic neuroendocrine tumors (GEP-NET). However, published data from Peru are limited. We hypothesize that this regimen could be a viable therapeutic option for advanced GEP-NET in the Peruvian population. AIM To evaluate overall survival (OS) in patients diagnosed with advanced GEP-NET treated with TEMCAP at the Instituto Nacional de Enfermedades Neoplásicas (INEN) in Lima-Perú. METHODS A retrospective review was conducted to identify patients with GEP-NEN treated with the TEMCAP regimen between 2011 and 2021 at the INEN. A total of thirty-eight patients were included in the final analysis: Thirty-five received TEMCAP as a first-line treatment, and three as a second-line treatment. The primary objective was to evaluate OS. The efficacy and safety of TEMCAP were assessed until the occurrence of unacceptable toxicity or disease progression. Survival outcomes were estimated using the Kaplan-Meier method. RESULTS The median age of the patients was 52 years (range 24-77 years), and 53.3% were female. The most common symptoms at diagnosis were abdominal pain in 31 patients (81.6%). Primary tumors included 12 in the rectum (31.6%), 11 in the pancreas (28.9%), 3 in the ileum (7.9%), 2 in the mesentery (5.3%), 2 in the small intestine (5.3%), 1 in the appendix (2.6%), 1 in the stomach (2.6%) and 6 cases of liver metastasis of unknown primary (15.8%). Five were neuroendocrine tumors (NET) G1 (13.2%), 33 were NET G2 (86.8%), five had Ki67 < 3% (13.2%), and 33 had Ki67 between 3% and 20% (86.8%). TEMCAP was administered to 35 (92.1%) patients as first-line treatment. OS at 12, 36, and 60 months was estimated in 80%, 66%, and 42%, respectively, with a median OS of 49 months. CONCLUSION TEMCAP therapy is a viable first-line option regarding efficacy and tolerability in areas where standard therapy is inaccessible.
Collapse
Affiliation(s)
| | - Victor Paitan
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Jersinho Medina
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Raymundo Flores
- Department of Radiology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Juan Haro-Varas
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Raul Mantilla
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Victor Castro-Oliden
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| |
Collapse
|