1
|
Millapán T, Gutiérrez Á, Rosas K, Buchegger K, Ili CG, Brebi P. In Silico Insights Reveal Fibronectin 1 as a Theranostic Marker in Gastric Cancer. Int J Mol Sci 2024; 25:11113. [PMID: 39456895 PMCID: PMC11507984 DOI: 10.3390/ijms252011113] [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: 09/28/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Gastric cancer (GC) is a complex and highly variable disease, ranking among the top five cancers diagnosed globally, and a leading cause of cancer-related deaths. Emerging from stomach lining cells amid chronic inflammation, it often advances to preneoplastic stages. Late-stage diagnoses and treatment challenges highlight the critical need for early detection and innovative biomarkers, motivating this study's focus on identifying theranostic markers through gene ontology analysis. By exploring deregulated biological processes, this study aims to uncover insights into cancer progression and associated markers, potentially identifying novel theranostic candidates in GC. Using public data from The Human Protein Atlas, this study pinpointed 299 prognostic genes, delineating 171 with unfavorable prognosis and 128 with favorable prognosis. Functional enrichment and protein-protein interaction analyses, supported by RNAseq results and conducted via Metascape and Cytoscape, highlighted five genes (vWF, FN1, THBS1, PCDH7, and F5) with promising theranostic potential. Notably, FN1 and THBS1 exhibited significant promise, with FN1 showing a 370% expression increase in cancerous tissue, and it is possible that FN1 can also indicate the stratification status in GC. While further validation is essential, these findings provide new insights into molecular alterations in GC and potential avenues for clinical application of theranostic markers.
Collapse
Affiliation(s)
- Tatiana Millapán
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Álvaro Gutiérrez
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Doctoral Program in Sciences with a Specialization in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4810296, Chile
| | - Krisnna Rosas
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Biotechnology Engineering Program, Universidad de La Frontera, Temuco 4810296, Chile
| | - Kurt Buchegger
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4810296, Chile
| | - Carmen Gloria Ili
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
| | - Priscilla Brebi
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (T.M.)
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- BMRC, Biomedical Research Consortium, Santiago 8331150, Chile
| |
Collapse
|
2
|
Smith TAD. Gene Abnormalities and Modulated Gene Expression Associated with Radionuclide Treatment: Towards Predictive Biomarkers of Response. Genes (Basel) 2024; 15:688. [PMID: 38927624 PMCID: PMC11202453 DOI: 10.3390/genes15060688] [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: 05/01/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Molecular radiotherapy (MRT), also known as radioimmunotherapy or targeted radiotherapy, is the delivery of radionuclides to tumours by targeting receptors overexpressed on the cancer cell. Currently it is used in the treatment of a few cancer types including lymphoma, neuroendocrine, and prostate cancer. Recently reported outcomes demonstrating improvements in patient survival have led to an upsurge in interest in MRT particularly for the treatment of prostate cancer. Unfortunately, between 30% and 40% of patients do not respond. Further normal tissue exposure, especially kidney and salivary gland due to receptor expression, result in toxicity, including dry mouth. Predictive biomarkers to select patients who will benefit from MRT are crucial. Whilst pre-treatment imaging with imaging versions of the therapeutic agents is useful in demonstrating tumour binding and potentially organ toxicity, they do not necessarily predict patient benefit, which is dependent on tumour radiosensitivity. Transcript-based biomarkers have proven useful in tailoring external beam radiotherapy and adjuvant treatment. However, few studies have attempted to derive signatures for MRT response prediction. Here, transcriptomic studies that have identified genes associated with clinical radionuclide exposure have been reviewed. These studies will provide potential features for seeding multi-component biomarkers of MRT response.
Collapse
Affiliation(s)
- Tim A D Smith
- Nuclear Futures Institute, School of Computer Science and Engineering, Bangor University, Dean Street, Bangor LL57 1UT, UK
| |
Collapse
|
3
|
Baum RP, Fan X, Jakobsson V, Yu F, Schuchardt C, Chen X, Zhang J. Long-term Nephrotoxicity after PRRT: Myth or Reality. Theranostics 2024; 14:451-459. [PMID: 38169589 PMCID: PMC10758070 DOI: 10.7150/thno.92487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale: The kidneys are commonly considered as the potential dose-limiting organ for peptide receptor radionuclide therapy (PRRT), making the risk of nephrotoxicity a primary concern. This retrospective analysis with prospective documentation and long-term follow-up aims to assess the risk of nephrotoxicity after PRRT in a large cohort of patients with neuroendocrine neoplasms (NENs) treated at our institution over the past 18 years. Methods: A total of 1361 NEN patients treated with 1-10 cycles of 177Lu-DOTA-TOC/-NOC/-TATE, 90Y-DOTA-TOC/-NOC/-TATE, DUO-PRRT (sequential administration of 90Y- and 177Lu-), or TANDEM-PRRT (combination of 90Y- and 177Lu- on the same day concomitantly) were included in this analysis. All parameters were prospectively documented in a structured database comprising over 250 items per patient and retrospectively analyzed. Kidney function, including serum creatinine, blood urea nitrogen, cGFR, and electrolytes, was evaluated before each PRRT cycle and during follow-up. Restaging was regularly performed at 6-month intervals until death. Treatment-related adverse events were graded according to the Common Terminology Criteria for Adverse Events (CTCAE v.5.0). Results: Between 2000 and 2018, a total of 5409 cycles of PRRT were administered to 1361 NEN patients. Follow-up after complete treatment was available for 1281 patients receiving 4709 cycles of PRRT, with a median follow-up time of 69.2 months (interquartile range, 32.8-110.5 months) and a maximum follow-up time of 175 months. Baseline creatinine levels were normal in 1039/1281 (81.1%) subjects, while grade 1 (G1) renal insufficiency was present in 221/1281 (17.3%) prior to PRRT. G2 was present in 19/1281 (1.5%), and G3 in 2/1281 (0.2%). After treatment, the proportion of G3/G4 grade patients only increased from 0.2% to 0.7%. Mean creatinine levels increased from a baseline of 0.90 ± 0.30 to 1.01 ± 0.57 mg/L (80.0 ± 26.7 to 89.4 ± 50.8 μmol/L) after treatment. In our main analysis cohort of 1244 patients (4576 cycles), 200 patients experienced an increase in CTCAE creatinine grade. Age, number of treatment cycles, type of radionuclides, and length of follow-up time were the main factors affecting CTCAE creatinine grading after treatment. When comparing the subgroups treated with different radionuclides, the risk of nephrotoxicity after 90Y treatment alone and the 90Y/177Lu combination group was higher than after 177Lu treatment alone. In the 90Y treatment subgroup, the two significant risk factors for an increased CTCAE creatinine grade were identified to be age (≥60) and a long follow-up time. Conclusions: This retrospective analysis with prospective documentation in a large cohort of 1281 NEN patients receiving 4709 cycles of PRRT co-administered with renal protection, treated through the individualized approach at a single institution over 18 years, did not reveal any evidence of long-term PRRT-related renal toxicity. The results of our study suggest that with the use of proper renal protection, nephrotoxicity due to PRRT is more likely a myth than a reality.
Collapse
Affiliation(s)
- Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Xin Fan
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Christiane Schuchardt
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Pakula RJ, Scott PJH. Applications of radiolabeled antibodies in neuroscience and neuro-oncology. J Labelled Comp Radiopharm 2023; 66:269-285. [PMID: 37322805 DOI: 10.1002/jlcr.4049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Positron emission tomography (PET) is a powerful tool in medicine and drug development, allowing for non-invasive imaging and quantitation of biological processes in live organisms. Targets are often probed with small molecules, but antibody-based PET is expanding because of many benefits, including ease of design of new antibodies toward targets, as well as the very strong affinities that can be expected. Application of antibodies to PET imaging of targets in the central nervous system (CNS) is a particularly nascent field, but one with tremendous potential. In this review, we discuss the growth of PET in imaging of CNS targets, present the promises and progress in antibody-based CNS PET, explore challenges faced by the field, and discuss questions that this promising approach will need to answer moving forward for imaging and perhaps even radiotherapy.
Collapse
Affiliation(s)
- Ryan J Pakula
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Pham TM, Duong VD, Doan VD, Vo VT, Le VT. Design synthesis of Y-90 glass microspheres and study of their therapeutic effects on mouse liver cancer cell line Hep3B. CHEMOSPHERE 2022; 299:134431. [PMID: 35358564 DOI: 10.1016/j.chemosphere.2022.134431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
In this article, a system for synthesizing Y-90 glass microspheres (Y-90-GM) was successfully designed in the Da Lat nuclear reactor (Vietnam), and the therapeutic effects of Y-90-GM on mice liver cancer cell line Hep3B were studied. The effects of synthesis factors, including heating time, heating temperature, gas flow rate, sample conduit length and diameter, were investigated to establish the optimal parameters. The size and shape of Y-90-GM were checked by field emission scanning electron microscope, and the radioactivity measurement was performed on a dosimeter. The results indicated that the optimal conditions for the synthesis of Y-90-GM were determined as the heating temperature of 1600 °C, heating time of 2 h, conduit length and diameter of 50 cm and 3.6 cm, and gas/oxygen flow rate of 15 mph. The Y-90-GM samples obtained at the optimal parameters have a size of 18-30 μm with a density of 3.53 g cm-3 and a specific radioactivity of 630 mCi g-1. The results of the therapeutic study on mice liver cancer cell line Hep3B showed that after two weeks of treatment with Y-90-GM (1mCi/mouse), the tumor volume was reduced by about 30.7% and after 3 consecutive treatment cycles, the liver cancer tumor was completely reduced. It was demonstrated that Y-90-GM is promising radiopharmaceuticals in the treatment of liver cancer by the radioembolization method.
Collapse
Affiliation(s)
- Thanh Minh Pham
- Center for Research and Production of Radioisotope, Dalat Nuclear Research Institute, 01 Nguyen Tu Luc, Dalat, 670000, Viet Nam
| | - Van Dong Duong
- Center for Research and Production of Radioisotope, Dalat Nuclear Research Institute, 01 Nguyen Tu Luc, Dalat, 670000, Viet Nam
| | - Van-Dat Doan
- The Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 70000, Viet Nam
| | - Van Thanh Vo
- Department of Human and Animal Physiology, Ho Chi Minh City University of Education, Ho Chi Minh City, 70000, Viet Nam
| | - Van Thuan Le
- Center for Advanced Chemistry, Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, 55000, Viet Nam; The Faculty of Natural Sciences, Duy Tan University, 03 Quang Trung, Da Nang, 55000, Viet Nam.
| |
Collapse
|
6
|
Klussmeier A, Aurich S, Niederstadt L, Wiedenmann B, Grötzinger C. Secretin Receptor as a Target in Gastrointestinal Cancer: Expression Analysis and Ligand Development. Biomedicines 2022; 10:biomedicines10030536. [PMID: 35327338 PMCID: PMC8944975 DOI: 10.3390/biomedicines10030536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Secretin was originally discovered as a gastrointestinal peptide that stimulates fluid secretion from the pancreas and liver and delays gastric emptying. In disease, a secretin receptor (SCTR) was found to occur as a splice variant in gastrinoma and pancreatic adenocarcinoma. Overexpression of SCTR has been described for gastrinomas, carcinoid tumors of the lung and cholangiocarcinoma. SCTR therefore is considered a candidate target for molecular tumor imaging as well as for peptide receptor radioligand therapy (PRRT) in a number of oncological indications. The aim of this study was to characterize SCTR expression in esophageal and pancreatic cancer, demonstrating for the first time high SCTR overexpression in these tumor types. In total, 65 of 70 pancreatic ductal adenocarcinoma tissues stained strongly positive for SCTR in immunohistochemistry, as did most of the 151 esophageal cancer samples, with minor influence of grading in both entities. In addition, the aim of this study was to further delineate residues in human secretin that are critical for binding to and activation of human SCTR. For a potential development of short and metabolically stable analogs for clinical use, it was intended to probe the peptide for its capacity to incorporate deletions and substitutions without losing its affinity to SCTR. In a systematic approach, a library of 146 secretin variants containing single amino acid substitutions as well as truncations on either end was tested in β-arrestin2-GFP translocation and fluorescent ligand internalization assays employing high-content analysis, in cAMP assays which run in agonist and antagonist mode, and in radioligand binding. The main structural determinants of SCTR binding and activation were localized to the N-terminus, with His1, Asp3 being among the most sensitive positions, followed by Phe6, Thr7 and Leu10. Aminoterminal truncation caused a rapid decline in receptor activity and most of these variants proved to be partial agonists showing antagonistic properties. In this study, the most potent novel antagonist showed an IC50 of 309 ± 74 nM in the β-arrestin2-GFP translocation assay on human SCTR while remaining a weak partial agonist. Future studies will have to demonstrate the utility of further enhanced secretin analogues as tracers for in vivo imaging and therapy.
Collapse
Affiliation(s)
- Anja Klussmeier
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
- Institut für Chemie und Biochemie, Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stefan Aurich
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Lars Niederstadt
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
- Partner Site Berlin, German Cancer Consortium (DKTK), 13353 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
7
|
Naik M, Al-Nahhas A, Khan SR. Treatment of Neuroendocrine Neoplasms with Radiolabeled Peptides-Where Are We Now. Cancers (Basel) 2022; 14:761. [PMID: 35159027 PMCID: PMC8833798 DOI: 10.3390/cancers14030761] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been one of the most successful and exciting examples of theranostics in nuclear medicine in recent decades and is now firmly embedded in many treatment algorithms for unresectable or metastatic neuroendocrine neoplasms (NENs) worldwide. It is widely considered to be an effective treatment for well- or moderately differentiated neoplasms, which express high levels of somatostatin receptors that can be selectively targeted. This review article outlines the scientific basis of PRRT in treatment of NENs and describes its discovery dating back to the early 1990s. Early treatments utilizing Indium-111, a γ-emitter, showed promise in reduction in tumor size and improvement in biochemistry, but were also met with high radiation doses and myelotoxic and nephrotoxic effects. Subsequently, stable conjugation of DOTA-peptides with β-emitting radionuclides, such as Yttrium-90 and Lutetium-177, served as a breakthrough for PRRT and studies highlighted their potential in eliciting progression-free survival and quality of life benefits. This article will also elaborate on the key trials which paved the way for its approval and will discuss therapeutic considerations, such as patient selection and administration technique, to optimize its use.
Collapse
Affiliation(s)
- Mitesh Naik
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| | | | - Sairah R. Khan
- Department of Imaging, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK;
| |
Collapse
|
8
|
Y90 selective internal radiation therapy and peptide receptor radionuclide therapy for the treatment of metastatic neuroendocrine tumors: combination or not? Nucl Med Commun 2021; 41:1242-1249. [PMID: 32941405 DOI: 10.1097/mnm.0000000000001284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Peptide receptor radionuclide therapy and selective internal radiation therapy are effective radionuclide therapy modalities for unresectable metastatic neuroendocrine tumor patients that cannot be controlled with somatostatin analogs. The present study is intended to evaluate the therapeutic efficacy and toxicity of the combined therapy of selective internal radiation therapy and peptide receptor radionuclide therapy and stand-alone selective internal radiation therapy in patients with neuroendocrine tumor, a liver-dominant disease. METHODS This cohort consists of 27 patients with metastatic neuroendocrine tumor and liver-dominant disease. They were grouped as the patients who were treated with selective internal radiation therapy for unresectable liver metastasis (n = 15) and the patients who received a combination of selective internal radiation therapy and peptide receptor radionuclide therapy (n = 12) for hepatic and extrahepatic metastasis. Treatment efficacy and treatment-associated toxicity were retrospectively assessed in both groups. RESULTS The objective treatment response and stable disease were found in 13 patients (86.6%) in the selective internal radiation therapy group and eight patients (66.6%) in the selective internal radiation therapy + peptide receptor radionuclide therapy group. The median overall survival rate was found to be 34.9 months, in the selective internal radiation therapy group and 67.5 months in the selective internal radiation therapy + peptide receptor radionuclide therapy group (P = 0.217). The median progression-free survival data was not reached, and the mean values of progression-free survival were 53.1 ± 9.9 months in the selective internal radiation therapy group, and 27.2 ± 5.9 months in the selective internal radiation therapy + peptide receptor radionuclide therapy group (P = 0.561). Temporary lymphopenia was the most common side effect. Grade 1-2 hepatotoxicity was observed to be 6.6% in the selective internal radiation therapy group, while it was not observed in selective internal radiation therapy + peptide receptor radionuclide therapy group. CONCLUSIONS In the neuroendocrine tumors with liver-dominant metastatic disease, personalized selective internal radiation therapy and peptide receptor radionuclide therapy and their combinations result in increased survival rates. Selective internal radiation therapy alone could be an effective treatment in patients with liver-limited and -dominant disease.
Collapse
|
9
|
Vahidfar N, Eppard E, Farzanehfar S, Yordanova A, Fallahpoor M, Ahmadzadehfar H. An Impressive Approach in Nuclear Medicine: Theranostics. PET Clin 2021; 16:327-340. [PMID: 34053577 DOI: 10.1016/j.cpet.2021.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Radiometal-based theranostics or theragnostics, first used in the early 2000s, is the combined application of diagnostic and therapeutic agents that target the same molecule, and represents a considerable advancement in nuclear medicine. One of the promising fields related to theranostics is radioligand therapy. For instance, the concepts of targeting the prostate-specific membrane antigen (PSMA) for imaging and therapy in prostate cancer, or somatostatin receptor targeted imaging and therapy in neuroendocrine tumors (NETs) are part of the field of theranostics. Combining targeted imaging and therapy can improve prognostication, therapeutic decision-making, and monitoring of the therapy.
Collapse
Affiliation(s)
- Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elisabeth Eppard
- Positronpharma SA, Santiago, Chile; Department of Nuclear Medicine, University Hospital Magdeburg, Germany
| | - Saeed Farzanehfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Fallahpoor
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
10
|
Ghaderi F, Jokar N, Gholamrezanezhad A, Assadi M, Ahmadzadehfar H. Toward radiotheranostics in cancer stem cells: a promising initial step for tumour eradication. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00444-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
11
|
Burkett BJ, Dundar A, Young JR, Packard AT, Johnson GB, Halfdanarson TR, Eiring RA, Gansen DN, Patton CM, Kendi AT. How We Do It: A Multidisciplinary Approach to 177Lu DOTATATE Peptide Receptor Radionuclide Therapy. Radiology 2020; 298:261-274. [PMID: 33231532 DOI: 10.1148/radiol.2020201745] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lutetium 177 (177Lu) DOTA-0-Tyr3-Octreotate (DOTATATE) peptide receptor radionuclide therapy (PRRT) is an effective treatment for advanced gastroenteropancreatic neuroendocrine tumors. This review presents a clinical practice workflow that has been successful since 177Lu DOTATATE PRRT was approved by the U.S. Food and Drug Administration. The workflow relies heavily on the input of a multidisciplinary team and involves a nuclear medicine consultation service, tumor board, and specific preparations in advance of therapy and day-of-therapy procedures. A systematic checklist designed to ensure appropriate selection of treatment candidates and identification of any concerns to address to safely administer PRRT is provided. All patients were evaluated with gallium 68 DOTATATE PET/CT, and in cases of high-grade tumors, they were also evaluated with fluorine 18 fluorodeoxyglucose PET/CT, with imaging findings reviewed as part of the systematic checklist before PRRT. Adverse effects are discussed and imaging follow-up regimens are reviewed, including alternative diagnostic contrast materials. Approaches to multiple challenging patient scenarios are illustrated through case examples. Finally, alternative theranostic radionuclides and treatment strategies are discussed.
Collapse
Affiliation(s)
- Brian J Burkett
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Ayca Dundar
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Jason R Young
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Annie T Packard
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Geoffrey B Johnson
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Thorvardur R Halfdanarson
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Rachel A Eiring
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Denise N Gansen
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - Cynthia M Patton
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| | - A Tuba Kendi
- From the Division of Nuclear Medicine, Department of Radiology (B.J.B., A.D., J.R.Y., A.T.P., G.B.J., D.N.G., C.M.P., A.T.K.), and Department of Medical Oncology (T.R.H., R.A.E.), Mayo Clinic, 200 First St SW, Rochester, MN 55905
| |
Collapse
|
12
|
Twelve-Year Survival of a Patient With Lymph Node, Pulmonary, Bone, Cardiac and Intraspinal Metastases of a Rectal Neuroendocrine Neoplasm Treated With Peptide Receptor Radionuclide Therapy-The Value of Salvage Peptide Receptor Radionuclide Therapy. Clin Nucl Med 2020; 45:e198-e200. [PMID: 31876836 DOI: 10.1097/rlu.0000000000002905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We report here the 12-year survival after the first peptide receptor radionuclide therapy (PRRT) of a patient with metastatic rectal neuroendocrine neoplasms, who received 7 cycles of PRRT with Lu/Y-DOTATATE/DOTATOC in 4 treatment phases. The patient demonstrated excellent response to each cycle of treatment, without any adverse effect even after repeated PRRT cycles. Most recently, immunohistochemistry revealed a G3 neuroendocrine neoplasm and intraspinal metastasis were successfully resected by neurosurgical intervention. This case nicely demonstrates that several "salvage" PRRTs can be given over many years leading to repetitive benefit for the patient and saving patients of possible toxicity of alternative treatments.
Collapse
|
13
|
Bajwa R, Madoff DC, Kishore SA. Embolotherapy for Hepatic Oncology: Current Perspectives and Future Directions. DIGESTIVE DISEASE INTERVENTIONS 2020; 4:134-147. [PMID: 32832829 DOI: 10.1055/s-0040-1712146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractPrimary and secondary liver cancers are a major cause of mortality worldwide. Transarterial liver-directed therapy, or embolotherapy, represents an important locoregional treatment strategy for primary and secondary liver tumors. Embolotherapeutic modalities include bland embolization (transarterial embolization), chemoembolization (transarterial chemoembolization), and radioembolization or selective internal radiotherapy. A brief technical overview of embolotherapeutic modalities as well as supportive evidence for the treatment of most common primary and secondary liver tumors will be discussed in this review. Several potential future applications, including synergy with systemic therapy, interventional theranostics, and artificial intelligence will also be reviewed briefly.
Collapse
Affiliation(s)
- Raazi Bajwa
- Memorial Sloan Kettering Cancer Center, Department of Radiology, Division of Interventional Radiology, New York, NY, USA
| | - David C Madoff
- Yale University School of Medicine, Department of Radiology and Biomedical Imaging, Division of Interventional Radiology, New Haven, CT, USA
| | - Sirish A Kishore
- Memorial Sloan Kettering Cancer Center, Department of Radiology, Division of Interventional Radiology New York, NY, USA
| |
Collapse
|
14
|
Peptide Receptor Radionuclide Therapy Using 225Ac-DOTATOC Achieves Partial Remission in a Patient With Progressive Neuroendocrine Liver Metastases After Repeated β-Emitter Peptide Receptor Radionuclide Therapy. Clin Nucl Med 2020; 45:241-243. [PMID: 31977472 DOI: 10.1097/rlu.0000000000002915] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We present here a case with β-radiation-refractory metastatic neuroendocrine tumors, who demonstrated an excellent therapy response after 1 cycle of Ac-DOTATOC, without any significant adverse effects even after 10 cycles of β-emitter peptide receptor radionuclide therapy followed by α-peptide receptor radionuclide therapy.
Collapse
|
15
|
Patient Selection and Toxicities of PRRT for Metastatic Neuroendocrine Tumors and Research Opportunities. Curr Treat Options Oncol 2020; 21:25. [PMID: 32172368 DOI: 10.1007/s11864-020-0711-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OPINION STATEMENT Neuroendocrine tumors (NETs) are a heterogenous group of neoplasms characterized by varied biological hallmarks and behavior, ranging from indolent to aggressive. For many decades, somatostatin analogues and few targeted therapies were available for NETs and these therapies had minimal response rates. However, there have been a number of recent treatment advances. Peptide receptor radionuclide therapy (PRRT) is a novel approach to treatment of NETs and has changed the landscape of treatment for NETs. It is a form of targeted therapy in which a radiolabeled somatostatin analogue delivers radiation specifically to tumor cells expressing the somatostatin receptor.
Collapse
|
16
|
Zhang J, Liu Q, Singh A, Schuchardt C, Kulkarni HR, Baum RP. Prognostic Value of 18F-FDG PET/CT in a Large Cohort of Patients with Advanced Metastatic Neuroendocrine Neoplasms Treated with Peptide Receptor Radionuclide Therapy. J Nucl Med 2020; 61:1560-1569. [PMID: 32169914 DOI: 10.2967/jnumed.119.241414] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/05/2020] [Indexed: 01/22/2023] Open
Abstract
The objective of this retrospective study was to determine the role of 18F-FDG PET/CT in a large cohort of 495 patients with metastatic neuroendocrine neoplasms (NENs) who were treated with peptide receptor radionuclide therapy (PRRT) with a long-term follow-up. Methods: The 495 patients were treated with 177Lu- or 90Y-DOTATOC/DOTATATE PRRT between February 2002 and July 2018. All subjects received both 68Ga-DOTATOC/TATE/NOC and 18F-FDG PET/CT before treatment and were followed 3-189 mo. Kaplan-Meier analysis, log-rank testing (Mantel-Cox), and Cox regression analysis were performed for overall survival (OS) and progression-free survival (PFS). Results: One hundred ninety-nine patients (40.2%) presented with pancreatic NENs, 49 with cancer of unknown primary, and 139 with midgut NENs, whereas the primary tumor was present in the rectum in 20, in the lung in 38, in the stomach in 8, and in other locations in 42. 18F-FDG PET/CT was positive in 382 (77.2%) patients and negative in 113 (22.8%) before PRRT, whereas 100% were 68Ga-DOTATOC/TATE/NOC-positive. For all patients, the median PFS and OS, defined from the start of PRRT, were 19.6 mo and 58.7 mo, respectively. Positive 18F-FDG results predicted shorter PFS (18.5 mo vs. 24.1 mo; P = 0.0015) and OS (53.2 mo vs. 83.1 mo; P < 0.001) than negative 18F-FDG results. Among the cases of pancreatic NENs, the median OS was 52.8 mo in 18F-FDG-positive subjects and 114.3 mo in 18F-FDG-negative subjects (P = 0.0006). For all patients positive for 18F-FDG uptake, and a ratio of more than 2 for the highest SUVmax on 68Ga-somatostatin receptor (SSTR) PET to the most 18F-FDG-avid tumor lesions, the median OS was 53.0 mo, compared with 43.4 mo in those patients with a ratio of less than 2 (P = 0.030). For patients with no 18F-FDG uptake (complete mismatch imaging pattern), the median OS was 108.3 mo versus 76.9 mo for an SUVmax of more than 15.0 and an SUVmax of 15.0 or less on 68Ga-SSTR PET/CT, respectively. Conclusion: The presence of positive lesions on 18F-FDG PET is an independent prognostic factor in patients with NENs treated with PRRT. Metabolic imaging with 18F-FDG PET/CT complements the molecular imaging aspect of 68Ga-SSTR PET/CT for the prognosis of survival after PRRT. High SSTR expression combined with negative 18F-FDG PET/CT results is associated with the most favorable long-term prognosis.
Collapse
Affiliation(s)
- Jingjing Zhang
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Qingxing Liu
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany.,Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; and
| | - Aviral Singh
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany.,GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Christiane Schuchardt
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Harshad R Kulkarni
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Richard P Baum
- Theranostics Center for Molecular Radiotherapy and Precision Oncology, ENETS Center of Excellence, Zentralklinik Bad Berka, Bad Berka, Germany
| |
Collapse
|
17
|
Delpassand ES, Ranganathan D, Wagh N, Shafie A, Gaber A, Abbasi A, Kjaer A, Tworowska I, Núñez R. 64Cu-DOTATATE PET/CT for Imaging Patients with Known or Suspected Somatostatin Receptor-Positive Neuroendocrine Tumors: Results of the First U.S. Prospective, Reader-Masked Clinical Trial. J Nucl Med 2020; 61:890-896. [PMID: 31924723 DOI: 10.2967/jnumed.119.236091] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/03/2020] [Indexed: 02/02/2023] Open
Abstract
Studies demonstrate that the investigational 64Cu-DOTATATE radiopharmaceutical may provide diagnostic and logistical benefits over available imaging agents for patients with somatostatin receptor (SSTR)-positive neuroendocrine tumors (NETs). Accordingly, we aimed to prospectively determine the lowest dose of 64Cu-DOTATATE that facilitates diagnostic-quality scans and evaluated the diagnostic performance and safety in a phase III study of patients with SSTR-expressing NETs. Methods: A dose-ranging study was conducted on 12 patients divided into 3 dose groups (111 MBq [3.0 mCi], 148 MBq [4.0 mCi], and 185 MBq [5.0 mCi] ± 10%) to determine the lowest dose of 64Cu-DOTATATE that produced diagnostic-quality PET/CT images. Using the 64Cu-DOTATATE dose identified in the dose-ranging study, 3 independent nuclear medicine physicians who were masked to all clinical information read PET/CT scans from 21 healthy volunteers and 42 NET-positive patients to determine those with disease or no disease, as well as those with localized versus metastatic status. Masked-reader evaluations were compared with a patient-specific standard of truth, which was established by an independent oncologist who used all previously available pathology, clinical, and conventional imaging data. Diagnostic performance calculated for 64Cu-DOTATATE included sensitivity, specificity, negative predictive value, positive predictive value, and accuracy. Inter- and intrareader reliability, as well as ability to differentiate between localized and metastatic disease, was also determined. Adverse events were recorded from 64Cu-DOTATATE injection through 48 h after injection. Results: The dose-ranging study identified 148 MBq (4.0 mCi) as the optimal dose to obtain diagnostic-quality PET/CT images. After database lock, diagnostic performance from an initial majority read of the 3 independent readers showed a significant 90.9% sensitivity (P = 0.0042) and 96.6% specificity (P < 0.0001) for detecting NETs, which translated to a 100.0% sensitivity and 96.8% specificity after correcting for an initial standard-of-truth misread. Excellent inter- and intrareader reliability, as well as ability to distinguish between localized and metastatic disease, was also noted. No adverse events were related to 64Cu-DOTATATE, and no serious adverse events were observed. Conclusion: 64Cu-DOTATATE PET/CT is a safe imaging technique that provides high-quality and accurate images at a dose of 148 MBq (4.0 mCi) for the detection of somatostatin-expressing NETs.
Collapse
Affiliation(s)
- Ebrahim S Delpassand
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas.,RadioMedix, Inc., Houston, Texas; and
| | | | | | - Afshin Shafie
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | - Ayman Gaber
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | | | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | | | - Rodolfo Núñez
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| |
Collapse
|
18
|
Scherübl H, Raue F, Frank-Raue K. [Neuroendocrine tumors : Classification, clinical presentation and imaging]. Radiologe 2019; 59:952-960. [PMID: 31359091 DOI: 10.1007/s00117-019-0574-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Neuroendocrine tumors (NET) are a heterogeneous group of neoplasms found in all organs. They often present with characteristic clinical syndromes due to hormone hypersecretion. DIAGNOSTICS In addition to hormone diagnostics molecular-genetic work-up can play an important role. IMAGING Morphological imaging comprises ultrasound, endoscopy, computed tomography (CT) and magnetic resonance imaging (MRI) scans. Functional imaging of NET relies on radioligands that bind to specific receptors or transporters (Ga-68-DOTATATE-PET-CT, Tc-99-tektrotyd-SPECT/CT, F‑18-DOPA-PET/CT). THERAPY Somatostatin analogs either native or coupled to radionuclides are potent drugs for treating various neuroendocrine tumors. CONCLUSION The requirements of imaging are determined by clinical presentation, laboratory findings, tumor stage, the presence of a tumor syndrome and the need of a personalized systemic treatment modality.
Collapse
Affiliation(s)
- H Scherübl
- Klinik für Innere Medizin, Gastroenterologie, GI Onkologie und Infektiologie, Vivantes Klinikum Am Urban, Dieffenbachstraße 1, 10967, Berlin, Deutschland.
| | - F Raue
- Endokrinologisch, nuklearmedizinische Praxis, Brückenstr. 21, 69120, Heidelberg, Deutschland
| | - K Frank-Raue
- Endokrinologisch, nuklearmedizinische Praxis, Brückenstr. 21, 69120, Heidelberg, Deutschland
| |
Collapse
|
19
|
Desai GS, Pande P, Chhabra V, Shah RC, Jagannath P. Multimodality management, recurrence patterns, and long-term outcome of gastroenteropancreatic neuroendocrine neoplasms: Progress over 17 years. Indian J Gastroenterol 2019; 38:399-410. [PMID: 31802438 DOI: 10.1007/s12664-019-00957-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Many advances in the management of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) happened in the last two decades. This study highlights the progress in its management over 17 years, outcomes, recurrence patterns, and follow up protocols. METHODS This retrospective analysis of prospectively maintained database at a single tertiary center included GEP-NEN patients from January 2001 to August 2017. Management protocols were based on European Neuroendocrine Tumor Society guidelines. Recurrences were categorized as follows: localized nodal, regional, distant hepatic, or combined. Patients were divided into cohorts: cohort 1 (2001-2006), cohort 2 (2007-2011), and cohort 3 (2012-2017). Survival patterns were analyzed. RESULTS One hundred and ninety-two patients were included with 98 (51.04%) grade (G) 1, 64 (33.34%) G2, and 30 (15.63%) G3. One hundred and four (54.16%) underwent curative surgery (58 G1, 27 G2, and 19 G3). Overall follow up ranged from 3 to 276 months; 39 were lost to follow up. Ninety-six patients had recurrences: 44 regional + distant and 40 liver-limited recurrences. One-, 3-, and 5-year survivals show significant differences among different treatment groups (p < 0.05). Significant increase in curative resections, chemotherapy utilization, and reduced recurrences were noted in cohort 3. Curative (R0) resection offered 1- and 3-year overall survival of 93.3% and 66.7% in cohort 1; 95.8% and 83.1% in cohort 2; and 100% and 92.9% in cohort 3. CONCLUSION Curative resection is the most significant factor for improved survival. Debulking surgerical procedure have a role whereas upfront peptide receptor radionuclide therapy is questionable. Chemotherapy improves overall survival in inoperable/metastatic setting. Recurrence patterns indicate that a long-term follow up greater than 10 years is necessary.
Collapse
Affiliation(s)
- Gunjan S Desai
- Department of Gastrointestinal Surgery, Lilavati Hospital and Research Center, Mumbai, 400 050, India. .,Department of Gastrointestinal Surgery, MPCT Hospital, C 7, Budhyadev Mandir Marg, Sector 4, Sanpada, Navi Mumbai, 400 705, India.
| | - Prasad Pande
- Department of Gastrointestinal Surgery, Lilavati Hospital and Research Center, Mumbai, 400 050, India
| | | | - Rajiv C Shah
- Department of Surgical Oncology, Lilavati Hospital and Research Centre, Mumbai, 400 050, India
| | - Palepu Jagannath
- Department of Surgical Oncology, Lilavati Hospital and Research Centre, Mumbai, 400 050, India
| |
Collapse
|
20
|
|
21
|
Bailly C, Bodet-Milin C, Bourgeois M, Gouard S, Ansquer C, Barbaud M, Sébille JC, Chérel M, Kraeber-Bodéré F, Carlier T. Exploring Tumor Heterogeneity Using PET Imaging: The Big Picture. Cancers (Basel) 2019; 11:cancers11091282. [PMID: 31480470 PMCID: PMC6770004 DOI: 10.3390/cancers11091282] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 01/02/2023] Open
Abstract
Personalized medicine represents a major goal in oncology. It has its underpinning in the identification of biomarkers with diagnostic, prognostic, or predictive values. Nowadays, the concept of biomarker no longer necessarily corresponds to biological characteristics measured ex vivo but includes complex physiological characteristics acquired by different technologies. Positron-emission-tomography (PET) imaging is an integral part of this approach by enabling the fine characterization of tumor heterogeneity in vivo in a non-invasive way. It can effectively be assessed by exploring the heterogeneous distribution and uptake of a tracer such as 18F-fluoro-deoxyglucose (FDG) or by using multiple radiopharmaceuticals, each providing different information. These two approaches represent two avenues of development for the research of new biomarkers in oncology. In this article, we review the existing evidence that the measurement of tumor heterogeneity with PET imaging provide essential information in clinical practice for treatment decision-making strategy, to better select patients with poor prognosis for more intensive therapy or those eligible for targeted therapy.
Collapse
Affiliation(s)
- Clément Bailly
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
| | - Caroline Bodet-Milin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
| | - Mickaël Bourgeois
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
- Groupement d'Intérêt Public Arronax, 44800 Saint-Herblain, France
| | - Sébastien Gouard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
| | - Catherine Ansquer
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
| | - Matthieu Barbaud
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
| | | | - Michel Chérel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
- Groupement d'Intérêt Public Arronax, 44800 Saint-Herblain, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, 44800 Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, 44800 Saint-Herblain, France
| | - Thomas Carlier
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44093 Nantes, France.
- Nuclear Medicine Department, University Hospital, 44093 Nantes, France.
| |
Collapse
|
22
|
The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part I). Mol Diagn Ther 2019; 23:1-26. [PMID: 30411216 DOI: 10.1007/s40291-018-0366-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present era of precision medicine sees 'cancer' as a consequence of molecular derangements occurring at the commencement of the disease process, with morphologic changes happening much later in the process of tumorigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound, and magnetic resonance imaging (MRI), play an integral role in the detection of disease at a macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumorigenesis, and thus has the potential to play a key role in heralding the transition from the concept of 'one size fits all' to 'precision medicine'. Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called 'radiogenomics', which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning, and artificial intelligence (AI), the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, a majority of these require validation before clinical translation is possible. In this two-part review, we discuss the systematic collaboration across structural, anatomical, and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
|
23
|
Langbein T, Dathe W, Deuerling A, Baum RP. Efficacy of Detoxsan ® powder on diarrhea caused by gastrointestinal neuroendocrine tumors. World J Gastroenterol 2019; 25:2133-2143. [PMID: 31114139 PMCID: PMC6506581 DOI: 10.3748/wjg.v25.i17.2133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/27/2019] [Accepted: 03/30/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Patients with neuroendocrine tumors (NETs) of the gastrointestinal tract suffer frequently from chronic diarrhea. A well characterized medical advice containing zeolite (Detoxsan® powder) was applied to patients suffered from therapy-refractory diarrhea either by its frequency or by watery stool, despite receiving standard pharmacotherapy according to the guidelines for carcinoid syndrome and comorbidities. Detoxsan® powder acts as an adsorbent and might reduce significantly symptoms of diarrhea in patients suffering from NETs.
AIM To overcome the therapy-refractory diarrhea of patients with NETs by the zeolite containing medical advice Detoxsan® powder.
METHODS A total of 20 patients (12 female and 8 male) suffering from diarrhea either by its frequency or from watery stool caused by NETs were included. In each patient, the diagnosis had been confirmed by histology and somatostatin receptors expression proven by positron emission tomography/computed tomography using Ga-68-labeled somatostatin analogs. All patients received standard-of-care pharmacotherapy and were additionally given Detoxsan® powder as an extemporaneous drug containing 90% natural Cuban zeolite and 10% magnesium aspartate. Recommended daily dosage ranges between 3 g once to three times per day. Each day dose and bowel movements were documented by the patients themselves in a pre-defined table. Additionally to the bowel movements quantitative determinations of serotonin, urea, creatinine and single ions were performed within the serum of the patients by commercially available equipment used as a matter of routine in the clinic.
RESULTS All patients enrolled in this pilot study did not only suffer from NETs, but also from comorbidities and treatment-resistant diarrhea. There was insufficient control of diarrhea, most probably due to the secretion of hormones like serotonin produced by the slowly growing and highly differentiated NETs. All patients only took Detoxsan® powder as an antidiarrheal drug. In general, response effects need several days to become perceptible and require an intake of Detoxsan® powder for an extended time period or intermittently, if persisting stabilization of bowel movements could not be achieved. A correlation between NET grade, part and size of bowel resection and functionality of the tumor could not be demonstrated. Therefore, diarrhea seemed to be based on the metabolic activity of the well-differentiated NETs, which eventually led to treatment resistance. In summary, 14 out of the 20 patients (70%) declared to be very content with using Detoxsan® powder and observed a significant reduction of diarrhea, while the effective dose and intake period that resulted in a symptom relief varied individually.
CONCLUSION Detoxsan® powder is able to reduce significantly symptoms of NET-related diarrhea in the majority of patients. The duration of taking Detoxsan® powder and its dosage vary individually.
Collapse
Affiliation(s)
- Thomas Langbein
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka D-99437, Germany
| | - Wilfried Dathe
- Scientific Managment, Heck Bio-Pharma GmbH, Winterbach D-73650, Germany
| | - Anika Deuerling
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka D-99437, Germany
| | - Richard P Baum
- Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka, Bad Berka D-99437, Germany
| |
Collapse
|
24
|
Zhang J, Kulkarni HR, Singh A, Niepsch K, Müller D, Baum RP. Peptide Receptor Radionuclide Therapy in Grade 3 Neuroendocrine Neoplasms: Safety and Survival Analysis in 69 Patients. J Nucl Med 2018; 60:377-385. [PMID: 30115686 DOI: 10.2967/jnumed.118.215848] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/07/2018] [Indexed: 12/19/2022] Open
Abstract
To date, limited data are available concerning peptide receptor radionuclide therapy (PRRT) of grade 3 (G3) neuroendocrine neoplasms (NENs) with a Ki-67 proliferation index of greater than 20%. The purpose of this study was to analyze the long-term outcome, efficacy, and safety of PRRT in patients with somatostatin receptor (SSTR)-expressing G3 NENs. Methods: A total of 69 patients (41 men; age, 28-81 y) received PRRT with 177Lu- or 90Y-labeled somatostatin analogs (DOTATATE or DOTATOC). Twenty-two patients had radiosensitizing chemotherapy. Kaplan-Meier analysis was performed to calculate progression-free survival (PFS) and overall survival (OS), defined from the start of PRRT, including a subgroup analysis for patients with a Ki-67 index of less than or equal to 55% and a Ki-67 index of greater than 55%. Treatment response was evaluated according to RECIST 1.1 as well as molecular imaging criteria (European Organization for Research and Treatment of Cancer). Short- and long-term toxicity was documented (Common Terminology Criteria for Adverse Events, v 5.0) using a structured database (comprising >250 items per patient) and retrospectively analyzed. Results: Forty-six patients had pancreatic NENs, 11 had unknown primary cancer, 6 had midgut NENs, 3 had gastric NENs, and 3 had rectal NENs. The median follow-up was 94.3 mo. The median PFS was 9.6 mo, and the median OS was 19.9 mo. For G3 NENs with a Ki-67 index of less than or equal to 55% (n = 53), the median PFS was 11 mo and the median OS was 22 mo. Patients with a Ki-67 index of greater than 55% (n = 11) had a median PFS of 4 mo and a median OS of 7 mo. For patients with positive SSTR imaging but no 18F-FDG uptake, the median PFS was 24 mo and the median OS was 42 mo. A significant difference was found for both PFS and OS, with median PFS of 16 mo and 5 mo and median OS of 27 mo and 9 mo for an SUVmax of greater than 15.0 and an SUVmax of less than or equal to 15.0, respectively, on SSTR PET. In the group with 18F-FDG uptake scored as 3 or 4, the median PFS was 7.1 mo and the median OS was 17.2 mo. In the group with 18F-FDG uptake scored as 0-2, the median PFS was 24.3 mo and the median OS was 41.6 mo. PRRT was well tolerated by all patients; no grade 3 or grade 4 hematotoxicity occurred, and no clinically significant decline in renal function was observed. There was no hepatotoxicity. Conclusion: PRRT was tolerated well, without significant adverse effects, and was efficacious in G3 NENs; the clinical outcome was promising, especially in patients with a Ki-67 index of less than or equal to 55% and even in patients for whom chemotherapy had failed. Baseline 18F-FDG along with SSTR molecular imaging was useful for stratifying G3 NEN patients with high uptake on SSTR PET/CT and no or minor 18F-FDG avidity-a mismatch pattern that was associated with a better long-term prognosis.
Collapse
Affiliation(s)
- Jingjing Zhang
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Harshad R Kulkarni
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Aviral Singh
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Karin Niepsch
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Dirk Müller
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Richard P Baum
- THERANOSTICS Center for Molecular Radiotherapy and Precision Oncology, Zentralklinik Bad Berka, Bad Berka, Germany
| |
Collapse
|
25
|
Bartsch DK, Scherübl H. Neuroendocrine Tumors of the Gastrointestinal Tract. Visc Med 2017; 33:321-322. [PMID: 29177159 DOI: 10.1159/000481766] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Marburg, Marburg, Germany
| | - Hans Scherübl
- Department of Gastroenterology, Gastrointestinal Oncology and Infectious Diseases, Vivantes Klinikum Am Urban, Berlin, Germany
| |
Collapse
|