1
|
Leunissen DJG, Moonen L, von der Thüsen JH, den Bakker MA, Hillen LM, van Weert TJJ, Zur Hausen A, van den Bosch TPP, Lap LMV, Damhuis RA, Reynaert NL, van den Broek EC, Fernandez-Cuesta L, Foll M, Alcala N, Sexton-Oates A, Dingemans AMC, Speel EJM, Derks JL. Identification of Defined Molecular Subgroups on the Basis of Immunohistochemical Analyses and Potential Therapeutic Vulnerabilities of Pulmonary Carcinoids. J Thorac Oncol 2025; 20:451-464. [PMID: 39581377 DOI: 10.1016/j.jtho.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Multi-omic studies have identified three molecular separated pulmonary carcinoid (PC) subgroups (A1, A2, B) with distinctive mRNA expression profiles (e.g., orthopedia homeobox protein [OTP], achaete-scute homolog [ASCL1], and hepatocyte nuclear factor 1 homeobox A [HNF1A]). We aimed to establish an immunohistochemical (IHC) biomarker panel that enables subgroup identification, and assessment of its potential clinical relevance. METHODS All patients with resected pulmonary carcinoids (2003-2012) were identified from the Dutch Cancer/Pathology Registry, and tumors were revised. The IHC expression of OTP, ASCL1, and HNF1A was scored in a blinded fashion in a mRNA-profiled (n = 5 per subgroup) and national carcinoid cohort (N = 478). The expression of potential therapeutic targets (somatostatin receptor type 2a [SSTR2A] and delta-like canonical Notch ligand 3 [DLL3]) was assessed. Immunohistochemistry was assessed using H-scoring. RESULTS OTP, ASCL1, and HNF1A reported similar IHC and mRNA expression patterns in the matched primary samples. In the national cohort, IHC separated PCs into subgroups A1 (n = 224 [53%], OTPhigh-ASCL1high-HNF1Alow), A2 (n = 161 [38%], OTPhigh-ASCL1low-HNF1Ahigh), and B (n = 37 [9%], OTPlow-ASCL1low-HNF1Ahigh). In 12% of PCs, no distinct classification could be provided. Patients with A1 were enriched for older age (83% > 50 y), female individuals (83%), and peripheral location (55%) with low SSTR2A (median = 10) and high DLL3 (median = 52) expression. A2 included younger patients (34% < 40 y) and endobronchial/central (87%) tumors with high SSTR2A (median = 160), but low DLL3 (median 0) expression. Group B included more male individuals (59%) and recurrence was more frequent (19%) than in groups A1 (8%) and A2 (6%). Neuroendocrine cell hyperplasia was enriched in A1 (25%) compared with A2 (3%) and B (0%). CONCLUSIONS An OTP, ASCL1, and HNF1A IHC panel enables the identification of molecular-defined pulmonary carcinoid subgroups with distinct clinical phenotypes and diverging therapeutic vulnerabilities that require further prospective evaluation.
Collapse
Affiliation(s)
- Daphne J G Leunissen
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Laura Moonen
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Lisa M Hillen
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Tijmen J J van Weert
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Axel Zur Hausen
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lisa M V Lap
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ronald A Damhuis
- Department of Research and Development, Association of Comprehensive Cancer Centres, Utrecht, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | | | - Lynnette Fernandez-Cuesta
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organization (IARC/WHO), Lyon, France
| | - Matthieu Foll
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organization (IARC/WHO), Lyon, France
| | - Nicolas Alcala
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organization (IARC/WHO), Lyon, France
| | - Alexandra Sexton-Oates
- Rare Cancers Genomics Team (RCG), Genomic Epidemiology Branch (GEM), International Agency for Research on Cancer/World Health Organization (IARC/WHO), Lyon, France
| | - Anne-Marie C Dingemans
- GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ernst-Jan M Speel
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jules L Derks
- GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Dev ID, Puranik AD, Trikalinos NA, Girod BJ, Kim H, Prasad V. Theranostics in Lung Neuroendocrine Tumors. Semin Nucl Med 2025; 55:221-233. [PMID: 40055049 DOI: 10.1053/j.semnuclmed.2025.02.010] [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: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 03/17/2025]
Abstract
In the last 2 decades, there has been a noticeable increase in the incidence of neuroendocrine tumors, in part due to improved understanding of pathology and/or availability of more sensitive and accurate diagnostic tests. While gastrointestinal tract and pancreas are the most common sites of origin, lung neuroendocrine tumors (LNETs) are also frequently reported and need special considerations from diagnostic as well as therapeutic aspects. Radiopharmaceutical therapy (Theranostics) is a novel approach which utilizes a pair of diagnostic and therapeutic agents that share a common target on tumor sites. Precise treatment of the disease with minimum side effects is the principal aim of Theranostics. It's a known fact that somatostatin receptors (SSTR) are abundantly expressed in neuroendocrine tumors. With the advent of highly specific radiopharmaceuticals targeting SSTR receptors for both diagnosis as well as treatment and other targeted therapies, management of LNETs has become less challenging. Still, there exists significant ambiguity in relation to management of LNETs with a scope of novel diagnostic and therapeutic strategies to pitch in. This review focuses on the role of established evidence for Theranostics strategies in the management of LNETs and highlights the potential future role of newer targets which would be of promising value in addressing such rare and complex tumor biology.
Collapse
Affiliation(s)
- Indraja D Dev
- Department of Nuclear Medicine and Molecular Imaging, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Center, Homi Bhabha National Institute, Navi Mumbai, Maharashtra, India
| | - Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nikolaos A Trikalinos
- Internal Medicine /Medical Oncology, Washington University in St Louis and Siteman Cancer Center, St Louis, MO
| | - Bradley John Girod
- Nuclear Medicine, Radiology, University of Texas Southwestern, Dallas, United States
| | - Hyun Kim
- Department of Radiation Oncology, Washington University School of Medicine in St. Louis, St Louis, MO
| | - Vikas Prasad
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University in Saint Louis, St Louis, MO.
| |
Collapse
|
3
|
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
|
4
|
Kiesewetter-Wiederkehr B, Melhorn P, Scheuba C, Raderer M. [Current developments in the treatment of neuroendocrine tumors]. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:568-574. [PMID: 38649498 PMCID: PMC11208222 DOI: 10.1007/s00117-024-01303-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Well-differentiated neuroendocrine tumors (NET) are rare malignancies that are clinically very heterogeneous. Accordingly, their treatment is also complex and dependent on various factors. With currently available systemic therapies, the prognosis is often favorable. OBJECTIVES This article aims to provide an overview of current treatment strategies for NET, addressing the most important NET locations. METHODS The current European guidelines and further relevant literature on the treatment of NET were reviewed for this purpose. RESULTS The therapeutic spectrum for NET is extremely broad: For NET of the stomach/duodenum, appendix, and rectum, endoscopic or surgical resection is often sufficient, and metastatic tumors are rare. NET of the pancreas, small intestine and lung should also undergo potentially curative resection in the early stages. In the metastatic stage, locoregional treatments such as surgery and liver tumor embolization play a role. Major advances have been made in drug therapy, with somatostatin analogs (octreotide and lanreotide), an mTOR inhibitor (everolimus), and a tyrosine kinase inhibitor (sunitinib) being widely used. Peptide receptor radionuclide therapy (PRRT) is also an invaluable option. In some cases, classic chemotherapy is indicated. CONCLUSIONS Many effective therapies are now available for NET. It is important to select the right therapy at the right time for each patient through interdisciplinary management.
Collapse
Affiliation(s)
- Barbara Kiesewetter-Wiederkehr
- Klinische Abteilung für Onkologie, Universitätsklinik für Innere Medizin I, Medizinische Universität Wien, Waehringer Guertel 18-20, 1090, Wien, Österreich.
| | - Philipp Melhorn
- Klinische Abteilung für Onkologie, Universitätsklinik für Innere Medizin I, Medizinische Universität Wien, Waehringer Guertel 18-20, 1090, Wien, Österreich
| | - Christian Scheuba
- Universitätsklinik für Allgemeinchirurgie, Medizinische Universität Wien, Wien, Österreich
| | - Markus Raderer
- Klinische Abteilung für Onkologie, Universitätsklinik für Innere Medizin I, Medizinische Universität Wien, Waehringer Guertel 18-20, 1090, Wien, Österreich
| |
Collapse
|
5
|
Malandrino P, Feola T, Mikovic N, Cannavale G, Molfetta SD, Altieri B, Mancini C, Ferolla P, Colao A, Faggiano A. Radioligand Therapy in Patients with Lung Neuroendocrine Tumors: A Systematic Review on Efficacy and Safety. Semin Nucl Med 2024; 54:570-580. [PMID: 38811266 DOI: 10.1053/j.semnuclmed.2024.05.001] [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: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
Neuroendocrine neoplasms (NENs), arising from various sites, present therapeutic challenges. Radioligand therapy (RLT) is effective for unresectable/metastatic NENs with increased somatostatin receptor uptake. While evidence supports RLT's efficacy in midgut NETs, its role in lung NETs remains underexplored. Clinical guidelines place RLT as a third or fourth-line option in this setting. However, in the last years several studies investigated mainly retrospectively effectiveness and safety of RLT in lung NET. The aim of this review is to assess the efficacy and safety of RLT in patients with lung NETs. Following PRISMA guidelines, a systematic review of MEDLINE and EMBASE databases retrieved English articles until March 31, 2023. Inclusion criteria encompassed studies involving RLT in lung NETs with efficacy and safety assessments. Twenty-seven studies met the criteria, totaling 786 patients. The pooled analysis revealed a 25.6% objective response rate and 75.6% disease control rate. Median progression-free survival averaged 20 months, while overall survival averaged 45 months. Factors affecting response included tumor burden, prior treatments, 18F-FDG PET scan uptake, and histological variants. RLT exhibited manageable grade 1/2 adverse effects, predominantly hematological, with Lu177 demonstrating a more favorable profile than Y90. The findings support RLT's effectiveness in lung NETs, offering hope for advanced SSTR-positive patients. Although identifying predictive factors for response remains challenging, RLT retained efficacy even after prior therapies and typical carcinoids displayed a slightly better response than atypical ones. Prospective trials are imperative to establish RLT's definitive efficacy and its place in the therapeutic landscape for lung NETs.
Collapse
Affiliation(s)
- Pasqualino Malandrino
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Catania, Italy.
| | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, Italy; Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - Nevena Mikovic
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Cannavale
- Department of Clinical Medicine and Surgery, UOC Endocrinology Diabetology and Andrology - University of Naples Federico II, Naples, Italy
| | - Sergio Di Molfetta
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Camilla Mancini
- Unit of Andrology and Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Piero Ferolla
- NET Multidisciplinary Group, Umbria Regional Cancer Network, Perugia, Italy
| | - Annamaria Colao
- UNESCO Chair, Education for Health and Sustainable Development, Federico II University, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, European Neuroendocrine Tumor Society (ENETS) Center of Excellence, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Fath MA, Liu D, Ewald JT, Robles-Planells C, Tomanek-Chalkley AM, Graves SA, Howe JR, O’Dorisio TM, Rastogi P, Bellizzi AM, Sue O’Dorisio M, Menda Y, Spitz DR. Chemokine Receptor CXCR4 Radioligand Targeted Therapy Using 177Lutetium-pentixather for Pulmonary Neuroendocrine Cancers. Radiat Res 2024; 201:35-47. [PMID: 37989124 PMCID: PMC10896455 DOI: 10.1667/rade-23-00064.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023]
Abstract
Intermediate to high-grade lung neuroendocrine tumors (NETs; i.e., atypical carcinoid tumors) and neuroendocrine carcinomas (NECs) are currently difficult to cure. These tumors were found to express the CXCR4 G-protein coupled receptor that can be targeted with radioligands. PCR and flow cytometric analysis of lung NET and NEC cell lines using an anti-CXCR4 antibody demonstrated that all cell lines tested expressed CXCR4. PET/CT imaging with 68Galium-pentixafor in mouse xenografts of NETs and NECs verified tumor targeting that was blocked by a CXCR4 agonist. Clonogenic survival analysis demonstrated a more than additive enhancement of killing when 1 μM auranofin (a thioredoxin reductase inhibitor) was used as a radiosensitizer in combination with 177Lu-pentixather (10 μCi). DMS273 small cell lung cancer xenografts in female nude mice treated with 25 μCi/g 177Lu-pentixather induced inhibition of tumor growth and resulted in an increase in overall survival without causing unacceptable normal tissue toxicities. Immunohistochemical staining of 95 retrospective human samples (containing 90 small cell lung carcinomas) demonstrated 84% CXCR4 positivity. In a multivariable analysis of this cohort that included age, gender, stage, primary site, SSTR2 status, and CXCR4 status, Cox regression models determined that only distant metastasis at presentation (P < 0.01) and a CXCR4 H-score >30 (P = 0.04) were significantly associated with reduced survival. Prospective clinical testing of patient tumors identified CXCR4-positivity in 76% of 21 NECs, 67% of 15 lung NETs (including 8 of 10 atypical carcinoids), and 0% of 25 non-lung NETs (including 5 NETS G3s). These data support the hypothesis that CXCR4-targeted theranostics can be utilized effectively for select NETs and NECs.
Collapse
Affiliation(s)
- Melissa A. Fath
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Dijie Liu
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Jordan T. Ewald
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Claudia Robles-Planells
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Ann M. Tomanek-Chalkley
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Stephen A. Graves
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - James R. Howe
- Department of Surgery, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Thomas M. O’Dorisio
- Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Prerna Rastogi
- Department of Pathology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Andrew M. Bellizzi
- Department of Pathology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - M Sue O’Dorisio
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Yusuf Menda
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| |
Collapse
|
7
|
Zhu T, Hsu JC, Guo J, Chen W, Cai W, Wang K. Radionuclide-based theranostics - a promising strategy for lung cancer. Eur J Nucl Med Mol Imaging 2023; 50:2353-2374. [PMID: 36929181 PMCID: PMC10272099 DOI: 10.1007/s00259-023-06174-8] [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: 12/15/2022] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE This review aims to provide a comprehensive overview of the latest literature on personalized lung cancer management using different ligands and radionuclide-based tumor-targeting agents. BACKGROUND Lung cancer is the leading cause of cancer-related deaths worldwide. Due to the heterogeneity of lung cancer, advances in precision medicine may enhance the disease management landscape. More recently, theranostics using the same molecule labeled with two different radionuclides for imaging and treatment has emerged as a promising strategy for systemic cancer management. In radionuclide-based theranostics, the target, ligand, and radionuclide should all be carefully considered to achieve an accurate diagnosis and optimal therapeutic effects for lung cancer. METHODS We summarize the latest radiotracers and radioligand therapeutic agents used in diagnosing and treating lung cancer. In addition, we discuss the potential clinical applications and limitations associated with target-dependent radiotracers as well as therapeutic radionuclides. Finally, we provide our views on the perspectives for future development in this field. CONCLUSIONS Radionuclide-based theranostics show great potential in tailored medical care. We expect that this review can provide an understanding of the latest advances in radionuclide therapy for lung cancer and promote the application of radioligand theranostics in personalized medicine.
Collapse
Affiliation(s)
- Tianxing Zhu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
- Lingang Laboratory, Shanghai, 200031, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jingpei Guo
- Department of Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Weiyu Chen
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kai Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
8
|
Urso L, Nieri A, Uccelli L, Castello A, Artioli P, Cittanti C, Marzola MC, Florimonte L, Castellani M, Bissoli S, Porto F, Boschi A, Evangelista L, Bartolomei M. Lutathera® Orphans: State of the Art and Future Application of Radioligand Therapy with 177Lu-DOTATATE. Pharmaceutics 2023; 15:pharmaceutics15041110. [PMID: 37111596 PMCID: PMC10142322 DOI: 10.3390/pharmaceutics15041110] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Lutathera® is the first EMA- and FDA-approved radiopharmaceutical for radioligand therapy (RLT). Currently, on the legacy of the NETTER1 trial, only adult patients with progressive unresectable somatostatin receptor (SSTR) positive gastroenteropancreatic (GEP) neuroendocrine neoplasms (NET) can be treated with Lutathera®. Conversely, patients with SSTR-positive disease arising from outside the gastroenteric region do not currently have access to Lutathera® treatment despite several papers in the literature reporting the effectiveness and safety of RLT in these settings. Moreover, patients with well-differentiated G3 GEP-NET are also still “Lutathera orphans”, and retreatment with RLT in patients with disease relapse is currently not approved. The aim of this critical review is to summarize current literature evidence assessing the role of Lutathera® outside the approved indications. Moreover, ongoing clinical trials evaluating new possible applications of Lutathera® will be considered and discussed to provide an updated picture of future investigations.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Alberto Nieri
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Licia Uccelli
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
- Correspondence: ; Tel.: +39-053-232-6387
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Paolo Artioli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| | - Maria Cristina Marzola
- Department of Nuclear Medicine, PET/CT Centre, S. Maria della Misericordia Hospital, 45100 Rovigo, Italy;
| | - Luigia Florimonte
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.C.); (L.F.); (M.C.)
| | - Sergio Bissoli
- Nuclear Medicine Unit, AULSS1 Dolomiti, San Martino Hospital, 32100 Belluno, Italy; (P.A.); (S.B.)
| | - Francesca Porto
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy; (L.U.); (C.C.); (F.P.)
| | - Alessandra Boschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Laura Evangelista
- Department of Medicine DIMED, University of Padua, 35128 Padua, Italy;
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy; (A.N.); (M.B.)
| |
Collapse
|
9
|
Kim H, El-Khoury V, Schulte N, Zhan T, Betge J, Cousin L, Felli E, Pessaux P, Ogier A, Opitz O, Ku B, Ebert MP, Kwon YJ. Personalized functional profiling using ex-vivo patient-derived spheroids points out the potential of an antiangiogenic treatment in a patient with a metastatic lung atypical carcinoid. Cancer Biol Ther 2022; 23:96-102. [PMID: 35193475 PMCID: PMC8890433 DOI: 10.1080/15384047.2021.2021042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Lung carcinoids are neuroendocrine tumors representing 1 to 2% of lung cancers. This study outlines the case of a patient with a metastatic lung atypical carcinoid who presented with a pleural effusion and progression of liver metastases after developing resistance to conventional treatments. Personalized functional profiling (PFP), i.e. drug screening, was performed in ex-vivo spheroids obtained from the patient's liver metastasis to identify potential therapeutic options. The drug screening results revealed cediranib, an antiangiogenic drug, as a hit drug for this patient, from a library of 66 Food and Drug Administration (FDA)-approved and investigational drugs. Based on the PFP results and the reported evidence of clinical efficacy of bevacizumab and capecitabine combination in gastro-intestinal neuroendocrine tumors, this combination was given to the patient. Four months later, the pleural effusion and pleura carcinosis regressed and the liver metastasis did not progress. The patient experienced 2 years of a stable disease under the PFP-guided personalized treatment.
Collapse
Affiliation(s)
- Hichul Kim
- Early Discovery and Technology Development, Ksilink, Strasbourg, France.,Personalized Therapy Discovery, Department of Oncology, Luxembourg Institute of Health, Dudelange, Luxembourg
| | - Victoria El-Khoury
- Personalized Therapy Discovery, Department of Oncology, Luxembourg Institute of Health, Dudelange, Luxembourg.,Luxembourg Center of Neuropathology (Lcnp), Department of Oncology, Luxembourg Institute of Health, Dudelange, Luxembourg
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany.,Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (Dkfz), Heidelberg, Germany
| | - Loic Cousin
- Early Discovery and Technology Development, Ksilink, Strasbourg, France
| | - Emanuele Felli
- Department of Visceral and Digestive Surgery, Nouvel Hopital Civil, University Hospital of Strasbourg, Strasbourg, France.,IHU-Strasbourg, Institute of Image-guided Surgery, Strasbourg, France.,Inserm Institute of Viral and Liver Disease (Inserm U1110), Strasbourg, France
| | - Patrick Pessaux
- Department of Visceral and Digestive Surgery, Nouvel Hopital Civil, University Hospital of Strasbourg, Strasbourg, France.,IHU-Strasbourg, Institute of Image-guided Surgery, Strasbourg, France.,Inserm Institute of Viral and Liver Disease (Inserm U1110), Strasbourg, France
| | - Arnaud Ogier
- Early Discovery and Technology Development, Ksilink, Strasbourg, France
| | - Oliver Opitz
- Coordinating Unit for Digital Medicine Baden-Württemberg (Ktbw), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bosung Ku
- Central R&D Center, Medical & Bio Decision (MBD), Suwon, Republic of Korea
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Yong-Jun Kwon
- Early Discovery and Technology Development, Ksilink, Strasbourg, France.,Personalized Therapy Discovery, Department of Oncology, Luxembourg Institute of Health, Dudelange, Luxembourg
| |
Collapse
|
10
|
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: 1.7] [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
|
11
|
Araujo-Castro M, Pascual-Corrales E, Molina-Cerrillo J, Moreno Mata N, Alonso-Gordoa T. Bronchial Carcinoids: From Molecular Background to Treatment Approach. Cancers (Basel) 2022; 14:520. [PMID: 35158788 PMCID: PMC8833538 DOI: 10.3390/cancers14030520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
A better understanding of the genetic and molecular background of bronchial carcinoids (BCs) would allow a better estimation of the risk of disease progression and the personalization of treatment in cases of advanced disease. Molecular studies confirmed that lungs neuroendocrine tumors (NETs) and neuroendocrine carcinomas (NECs) are different entities; thus, no progression of NET to NEC is expected. In BCs, MEN1 gene mutations and deletions and decreased gene expression have been associated with a poor prognosis. ATRX mutation has also been linked to a shorter disease-specific survival. In terms of therapeutic targets, PI3K/AKT/mTOR pathway mutations have been described in 13% of typical carcinoids (TCs) and 39% of atypical carcinoids (ACs), representing a targetable mutation with kinase inhibitors. Regarding treatment, surgical resection is usually curative in localized BCs and adjuvant treatment is not routinely recommended. Multiple options for systemic therapy exist for patients with advanced BCs, although limited by a heterogeneity in the scientific evidence behind their use recommendation. These options include somatostatin analogues, everolimus, peptide receptor radionuclide therapy, chemotherapy, radiotherapy, antiangiogenic agents, and immunotherapy. In this article, we provide a comprehensive review about the molecular and genetic background of BCs, and about the treatment of local and metastatic disease, as well as the main paraneoplastic syndromes that have been associated with this tumor.
Collapse
Affiliation(s)
- Marta Araujo-Castro
- Neuroendocrinology Unit, Endocrinology and Nutrition Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
- Instituto de Investigación Biomédica Ramón y Cajal (IRICYS), 28034 Madrid, Spain;
- Universidad de Alcalá, 28801 Madrid, Spain
| | - Eider Pascual-Corrales
- Neuroendocrinology Unit, Endocrinology and Nutrition Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
- Instituto de Investigación Biomédica Ramón y Cajal (IRICYS), 28034 Madrid, Spain;
| | - Javier Molina-Cerrillo
- Instituto de Investigación Biomédica Ramón y Cajal (IRICYS), 28034 Madrid, Spain;
- Universidad de Alcalá, 28801 Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Nicolás Moreno Mata
- Thoracic Surgery Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
| | - Teresa Alonso-Gordoa
- Instituto de Investigación Biomédica Ramón y Cajal (IRICYS), 28034 Madrid, Spain;
- Universidad de Alcalá, 28801 Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| |
Collapse
|
12
|
Tong D, Zhang Y, Jiang J, Bi G. Identification of a VHL gene mutation in atypical Von Hippel-Lindau syndrome: genotype-phenotype correlation and gene therapy perspective. Cancer Cell Int 2021; 21:685. [PMID: 34923986 PMCID: PMC8684656 DOI: 10.1186/s12935-021-02386-w] [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: 12/21/2020] [Accepted: 12/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background Classical von Hippel Lindau (VHL) disease/syndrome includes CNS hemangioblastoma, renal or pancreatic cysts, pheochromocytoma, renal carcinoma and exodermic cystadenoma. The syndrome is caused by mutation of VHL tumor suppressor gene. The most prevalent mutations are present in VHL syndrome. To date, > 500 mutations of gene related to the progression of VHL syndrome have been reported. VHL gene mutation presented in single lung or pancreatic tumor has been reported occasionally, but there is no report of both. Methods In this paper, we used CT scan, pathological and genetic examination methods to diagnose a rare atypical VHL syndrome. Results We reported a rare case of atypical VHL syndrome with authenticated VHL mutation at p.Arg167Gln, that was associated with not only bilateral pheochromocytoma but also lung carcinoid and neuroendocrine tumor of pancreas. Based on literature reviews, the patient was recommended to be further subjected to octreotide-based radionuclide therapy. Conclusions Combined with gene detection and clinical diagnosis, we found the inherent relationship between VHL genotype and phenotype, and constructed the standard diagnosis and treatment process of disease with rare VHL mutation from the perspective of gene therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02386-w.
Collapse
Affiliation(s)
- Dali Tong
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Yao Zhang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| | - Gang Bi
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
13
|
Ramirez RA, Thomas K, Jacob A, Lin K, Bren-Mattison Y, Chauhan A. Adjuvant therapy for lung neuroendocrine neoplasms. World J Clin Oncol 2021; 12:664-674. [PMID: 34513600 PMCID: PMC8394158 DOI: 10.5306/wjco.v12.i8.664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary neuroendocrine neoplasms (NENs) represent a minority of lung cancers and vary from slower growing pulmonary carcinoid (PC) tumors to aggressive small cell lung cancer (SCLC). While SCLC can account for up to 15% of lung cancer, PCs are uncommon and represent about 2% of lung cancers. Surgical resection is the standard of care for early-stage PCs and should also be considered in early stage large cell neuroendocrine carcinoma (LCNEC) and SCLC. Adjuvant treatment is generally accepted for aggressive LCNEC and SCLC, however, less well established for PCs. Guidelines admit a lack of trials to support a high-level recommendation for adjuvant therapy. This manuscript will discuss the role for adjuvant therapy in NENs and review the available literature.
Collapse
Affiliation(s)
- Robert A Ramirez
- Department of Medicine-Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Katharine Thomas
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, United States
| | - Aasems Jacob
- Division of Medical Oncology, University of Kentucky, Lexington, KY 40536, United States
| | - Karen Lin
- Department of Oncology, Brookwood Baptist Health, Birmingham, AL 35209, United States
| | - Yvette Bren-Mattison
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112, United States
- New Orleans Louisiana Neuroendocrine Tumor Specialists (NOLANETS), Ochsner Medical Center, Kenner, LA 70065, United States
| | - Aman Chauhan
- Division of Medical Oncology, University of Kentucky, Lexington, KY 40536, United States
| |
Collapse
|
14
|
Abstract
Peptide receptor radionuclide therapy (PRRT) is an effective form of treatment of patients with metastatic neuroendocrine tumors, delivering modest objective tumor response rates but notable survival and symptomatic benefits. The first PRRT approved by the US Food and Drug Administration was lutetium 177-DOTATATE and is for use in adults with somatostatin receptor-positive gastroenteropancreatic neuroendocrine tumors. The treatment paradigm typically leads to significant improvement in symptomology coupled with an extended period of progression-free survival. Side effects are limited, with a small fraction of individuals experiencing clinically significant long-term renal or hematologic toxicity.
Collapse
|
15
|
Prantesh J, Dorth J, Asa SL, Mohamed A. Nasopharyngeal neuroendocrine neoplasms: Systematic review of the literature and case presentation. J Neuroendocrinol 2021; 33:e13005. [PMID: 34342078 DOI: 10.1111/jne.13005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/20/2021] [Accepted: 06/15/2021] [Indexed: 01/04/2023]
Abstract
Most of neuroendocrine neoplasms (NENs) are located in the gastrointestinal tract and lung, and they are rarely found on the upper aero-digestive tract, which limit the current literature about nasopharyngeal NENs. This systemic review will summarize the clinical, pathological features and optimal diagnosis and management of different types of nasopharyngeal NENs (NP NENs). In-addition, we herein report an EBV negative TP53-mutated/ Rb-wild type nasopharyngeal neuroendocrine carcinoma (NEC) in a young man in which touch preparation cytology studies were integral to establishing a definitive diagnosis. To our knowledge, only very few cases of primary neuroendocrine carcinoma of the nasopharynx have been reported in the literature and the reports of these cases have not included detailed description of different types and how to optimally diagnose and manage them. In this abstract, we also highlighted the evidence about the safety of using growth factors in patients with sickle cell anemia who are receiving cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Jain Prantesh
- Department of Medicine, Division of Hematology and Medical Oncology, Seidman Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Jennifer Dorth
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Sylvia L Asa
- Department of Pathology, Seidman Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Amr Mohamed
- Department of Medicine, Division of Hematology and Medical Oncology, Seidman Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
16
|
Altıparmak Güleç B, Yurt F. Treatment with Radiopharmaceuticals and Radionuclides in Breast Cancer: Current Options. Eur J Breast Health 2021; 17:214-219. [PMID: 34263148 DOI: 10.4274/ejbh.galenos.2021.2021-3-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/20/2021] [Indexed: 12/22/2022]
Abstract
Radiopharmaceutical therapy (RPT) is an effective and safe treatment for many types of cancer. RPT acts by binding radioactive atoms to tumor-associated antigens, monoclonal antibodies, nanoparticles, peptides, and small molecules. These treatments ensure that a concentrated dose is delivered to the targeted tumor tissue while preserving the normal tissues surrounding the tumor. Given these features, RPT is superior to traditional methods. This review article aimed to performa comprehensive review and evaluation of the potential of radionuclides and radiopharmaceuticals used in breast cancer treatment in preclinical studies conducted in the last five years.
Collapse
Affiliation(s)
- Burcu Altıparmak Güleç
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, İzmir, Turkey (Graduated)
| | - Fatma Yurt
- Department of Nuclear Applications, Institute of Nuclear Science, Ege University, İzmir, Turkey
| |
Collapse
|
17
|
Derks JL, Rijnsburger N, Hermans BCM, Moonen L, Hillen LM, von der Thüsen JH, den Bakker MA, van Suylen RJ, Speel EJM, Dingemans AMC. Clinical-Pathologic Challenges in the Classification of Pulmonary Neuroendocrine Neoplasms and Targets on the Horizon for Future Clinical Practice. J Thorac Oncol 2021; 16:1632-1646. [PMID: 34139363 DOI: 10.1016/j.jtho.2021.05.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022]
Abstract
Diagnosing a pulmonary neuroendocrine neoplasm (NEN) may be difficult, challenging clinical decision making. In this review, the following key clinical and pathologic issues and informative molecular markers are being discussed: (1) What is the preferred outcome parameter for curatively resected low-grade NENs (carcinoid), for example, overall survival or recurrence-free interval? (2) Does the WHO classification combined with a Ki-67 proliferation index and molecular markers, such as OTP and CD44, offer improved prognostication in low-grade NENs? (3) What is the value of a typical versus atypical carcinoid diagnosis on a biopsy specimen in local and metastatic disease? Diagnosis is difficult in biopsy specimens and recent observations of an increased mitotic rate in metastatic carcinoid from typical to atypical and high-grade NEN can further complicate diagnosis. (4) What is the (ir)relevance of morphologically separating large cell neuroendocrine carcinoma (LCNEC) SCLC and the value of molecular markers (RB1 gene and pRb protein or transcription factors NEUROD1, ASCL1, POU2F3, or YAP1 [NAPY]) to predict systemic treatment outcome? (5) Are additional diagnostic criteria required to accurately separate LCNEC from NSCLC in biopsy specimens? Neuroendocrine morphology can be absent owing to limited sample size leading to missed LCNEC diagnoses. Evaluation of genomic studies on LCNEC and marker studies have identified that a combination of napsin A and neuroendocrine markers could be helpful. Hence, to improve clinical practice, we should consider to adjust our NEN classification incorporating prognostic and predictive markers applicable on biopsy specimens to inform a treatment outcome-driven classification.
Collapse
Affiliation(s)
- Jules L Derks
- Department of Pulmonary Diseases, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Nicole Rijnsburger
- Department of Respiratory Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bregtje C M Hermans
- Department of Pulmonary Diseases, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Laura Moonen
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Lisa M Hillen
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Robert J van Suylen
- Pathology-DNA, Location Jeroen Bosch Hospital, s' Hertogenbosch, The Netherlands
| | - Ernst-Jan M Speel
- Department of Pathology, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonary Diseases, GROW School for Oncology & Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Respiratory Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Neuroendocrine Tumor Theranostics: An Update and Emerging Applications in Clinical Practice. AJR Am J Roentgenol 2021; 217:495-506. [PMID: 34076455 DOI: 10.2214/ajr.20.23349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE. Theranostics have shown great promise for delivering precision medicine, particularly in neuroendocrine tumors (NETs). The clinical applications of radiolabeled somatostatin analogues in imaging and radionuclide therapy have been rapidly increasing over the past 2 decades and are currently integrated into the management guidelines of NETs. This article summarizes the available literature on different somatostatin receptor-targeting radiopharmaceuticals with theranostic potential in NETs, pheochromocytomas, and paragangliomas. We discuss the clinical application, administration, and toxicity of recent FDA-approved radionuclide therapies, including 177Lu-DOTATATE in advanced gastroenteropancreatic NETs and 131I-MIBG in advanced paragangliomas and pheochromocytomas. CONCLUSION. Several studies support the safety and clinical efficacy of peptide receptor radionuclide therapies in disease control and quality-of-life improvement in patients with NETs and report potential benefits of combined radionuclide treatment approaches. The utility and pitfalls of functional imaging in therapy response assessment and surveillance of NETs remain to be established.
Collapse
|
19
|
Zidan L, Iravani A, Oleinikov K, Ben-Haim S, Gross DJ, Meirovitz A, Maimon O, Akhurst T, Michael M, Hicks RJ, Grozinsky-Glasberg S, Kong G. Efficacy and safety of 177Lu-DOTATATE in lung neuroendocrine tumors: a bi-center study. J Nucl Med 2021; 63:218-225. [PMID: 34049983 DOI: 10.2967/jnumed.120.260760] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/21/2021] [Indexed: 11/16/2022] Open
Abstract
To assess the efficacy and safety of 177Lu-DOTATATE in patients with somatostatin receptor (SSR) positive lung neuroendocrine tumor (NET). Methods: This is a retrospective review of the outcome of patients with typical carcinoid (TC) and atypical carcinoid (AC), treated with 177Lu-DOTATATE at two ENETS Centres of Excellence. Morphological imaging (RECIST 1.1) and 68Ga-DOTATATE PET/CT responses were assessed at 3 months after completion of 177Lu-DOTATATE. Concordance between two response assessment methods was evaluated by Kappa statistics. Progression-free survival (PFS) and overall survival (OS) was estimated by Kaplan-Meier analysis and compared by Log-rank test. Treatment-related adverse events (AEs) were graded based on CTCAE version 5. Results: Of 48 patients (median age, 63 years, 13 female), 43 (90%) had AC and 5 (10%) TC. Almost all patients (47, 98%) were treated due to progression. Majority (40, 83%) received somatostatin analogs and 10 patients (20%) had prior everolimus, chemotherapy or both. All patients had high SSR expression (≥ modified Krenning score 3) on pre-treatment 68Ga-DOTATATE PET/CT. Patients received a median 4 (range 1-4) cycles of 177Lu-DOTATATE (33% with concurrent radiosensitizing chemotherapy) to a median cumulative activity of 27GBq (range 6-43GBq). At median follow-up of 42 months, the median PFS and OS were 23 months (95% CI 18-28 months) and 59 months (95% CI 50-not reached [NR]), respectively. Of 40 patients with RECIST-measurable disease and 39 patients with available 68Ga-DOTATATE PET/CT response categories were: partial response, 20% (95% CI 10-35%) and 44% (95% CI 30-59%); stable disease, 68% (95% CI 52-80%) and 44% (95% CI 30-59%) and progressive disease 12% (95% CI 5-27%) by both, respectively. There was a moderate concordance between response categories by RECIST and 68Ga-DOTATATE PET/CT, weighted Kappa of 0.51 (95% CI 0.21-0.68). Of patients with stable disease by RECIST, those with partial response on 68Ga-DOTATATE PET/CT had longer OS compared to those with no response, NR vs 52 months (95% CI 28-64), HR 0.2 (95% CI 0.1-0.6), p 0.001. Most grade 3/4 AEs were reversible and the most common was lymphopenia (14%) with no incidence of myelodysplasia/leukemia. Conclusion: In patients with advanced progressive lung NET and satisfactory SSR expression, 177Lu-DOTATATE is effective and safe with a high disease control rate and encouraging PFS and OS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Grace Kong
- Peter MacCallum Cancer Centre, Australia
| |
Collapse
|
20
|
Indications of Peptide Receptor Radionuclide Therapy (PRRT) in Gastroenteropancreatic and Pulmonary Neuroendocrine Tumors: An Updated Review. J Clin Med 2021; 10:jcm10061267. [PMID: 33803817 PMCID: PMC8003169 DOI: 10.3390/jcm10061267] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Radionuclide therapy for neuroendocrine tumors is a form of systemic radiotherapy that allows the administration of targeted radionuclides into tumor cells that express a large quantity of somatostatin receptors. The two most commonly used radio-peptides for radionuclide therapy in neuroendocrine tumors are 90Y-DOTATOC and 177Lu-DOTATATE. Radio-peptides have been used for several years in the treatment of advanced neuroendocrine tumors. Recently, the randomized Phase III study NETTER-1 compared177Lu-DOTATATE versus high-dose (double-dose) octreotide LAR in patients with metastatic midgut neuroendocrine tumors, and demonstrated its efficacy in this setting. Strong signals in favor of efficiency seem to exist for other tumors, in particular for pancreatic and pulmonary neuroendocrine tumors. This focus on radionuclide therapy in gastroenteropancreatic and pulmonary neuroendocrine tumors addresses the treatment modalities, the validated and potential indications, and the safety of the therapy.
Collapse
|
21
|
Djekidel M, Syed G, Kanbour A. Imaging Biomarkers in Lung Cancer with 68Ga-DOTATATE, 18F-Fluoride, and 18F-FDG PET/CT Scans and the Theranostics Paradigm. J Nucl Med Technol 2021; 49:281-283. [PMID: 33722922 DOI: 10.2967/jnmt.120.258343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/27/2021] [Indexed: 11/16/2022] Open
Abstract
Lung cancer is the number 1 cause of cancer deaths in the United States. The prognosis is quite grim with the exception of stage 1. When faced with several failed therapeutic regimens and rapid progression of the disease, considering alternative therapies such as radiopharmaceutical therapies may be an option. We describe the case of a 36-y-old man with lung adenocarcinoma who had imaging molecular characterization of his disease with 18F-FDG, 68Ga-DOTATATE, and 18F-fluoride PET/CT scans that were able to shed some light on molecular characterization of his disease and serve as a guide to potential targeted or personalized radiopharmaceutical therapeutic options.
Collapse
Affiliation(s)
- Mehdi Djekidel
- NCCCR, Division of Oncology, Hamad Medical Corporation, Doha, Qatar;
| | - Ghulam Syed
- Department of Radiology, Division of Nuclear Medicine, Hamad Medical Corporation, Doha, Qatar; and
| | - Aladdin Kanbour
- Department of Radiology, Division of Nuclear Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
22
|
Snene H, Badri I, Mehiri N, Ben Salah N, Blibech H, Aouina H, Belhadj S, Boussen H, Chaouch N, Charfi R, Fenniche S, Gharbi L, Ghrairi H, Hamzaoui A, Megdiche L, Merai S, Mezni F, Tritar F, Daghfous J, Marghli A, Louzir B. [Diagnostic and therapeutic management of operable bronchopulmonary carcinoid tumours]. Rev Mal Respir 2021; 38:249-256. [PMID: 33674138 DOI: 10.1016/j.rmr.2021.02.062] [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: 07/28/2020] [Accepted: 02/10/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Bronchial carcinoid tumours (CT), divided into typical carcinoid (TC) or atypical carcinoid (AC), are rare tumours whose therapeutic management remains unspecified. METHODS Retrospective study collecting cases of bronchial CT operated at the thoracic surgery department of Abderrahmane-Mami hospital of Ariana and recruited from the pneumology departments of Northern Tunisia, during a 12-year period. RESULTS Ninety patients were collected (74 cases of TC and 16 cases of AC). The mean age was 45 years and the sex ratio H/F=0.5. The chest X-ray was normal in 11 cases, as well as flexible bronchoscopy in seven cases. The tumour was classified: stage IA (10 cases), IIA (28 cases), IIB (31 cases), IIIA (15 cases) and IIIB (six cases). Surgery resulted in a complete resection in 78 patients, an extensive resection in six patients, and a conservative resection in six patients. Adjuvant chemotherapy was given in 10 patients. The survival was 84% at five years and 42% at 10 years. CONCLUSION The prognosis of CT depends directly on the histological subtype. It is excellent for TC after complete resection, unlike ACs that are similar to well-differentiated bronchial carcinomas.
Collapse
Affiliation(s)
- H Snene
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie.
| | - I Badri
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| | - N Mehiri
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| | - N Ben Salah
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| | - H Blibech
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| | - H Aouina
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Charles-Nicolle, Tunis, Tunisie
| | - S Belhadj
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, hôpital régional de Menzel-Bourguiba, Bizerte, Tunisie
| | - H Boussen
- Faculté de médecine de Tunis, université de Tunis El Manar, service d'oncologie médicale, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - N Chaouch
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon 2, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - R Charfi
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie, hôpital des forces de sécurité intérieur, Tunis, Tunisie
| | - S Fenniche
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon 4, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - L Gharbi
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon D, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - H Ghrairi
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie, hôpital Taher-Maamouri, Nabeul, Tunisie
| | - A Hamzaoui
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon B, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - L Megdiche
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon Ibn Nafis, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - S Merai
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie, CHU La Rabta, Tunis, Tunisie
| | - F Mezni
- Faculté de médecine de Tunis, université de Tunis El Manar, service d'anatomie pathologie, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - F Tritar
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie pavillon C, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - J Daghfous
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| | - A Marghli
- Faculté de médecine de Tunis, université de Tunis El Manar, service de chirurgie thoracique et cardiovasculaire, CHU Abderrahmen-Mami, Ariana, Tunisie
| | - B Louzir
- Faculté de médecine de Tunis, université de Tunis El Manar, service de pneumologie allergologie, CHU Mongi-Slim La Marsa, 2047, Sidi-Daoued, Tunis, Tunisie
| |
Collapse
|
23
|
Haider M, Das S, Al-Toubah T, Pelle E, El-Haddad G, Strosberg J. Somatostatin receptor radionuclide therapy in neuroendocrine tumors. Endocr Relat Cancer 2021; 28:R81-R93. [PMID: 33608483 PMCID: PMC8118168 DOI: 10.1530/erc-20-0360] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lu-DOTATATE has been approved for the treatment of gastroenteropancreatic NETs. An understanding of benefits and risks is important for the appropriate implementation of this therapy. This review summarizes study data supporting the use of radiolabeled somatostatin analogs for the treatment of advanced NETs and highlights risks, including potential toxicities in specific populations. Key ongoing clinical trials, including randomized studies, are designed to better define the position of PRRT within the broader therapeutic landscape. Preclinical and early-phase human studies are focused on the development of novel somatostatin-receptor agonists and antagonists, new radionuclides, and radiosensitizing combination therapies.
Collapse
Affiliation(s)
- Mintallah Haider
- Moffitt Cancer Center, Department of GI Oncology, Tampa, Florida, USA
| | - Satya Das
- Department of GI Oncology, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Eleonora Pelle
- Department of Oncology, University of Bari, Bari, Puglia, Italy
| | - Ghassan El-Haddad
- Moffitt Cancer Center, Department of Diagnostic Imaging and Interventional Radiology, Tampa, Florida, USA
| | | |
Collapse
|
24
|
Sonbol MB, Halfdanarson TR, Hilal T. Assessment of Therapy-Related Myeloid Neoplasms in Patients With Neuroendocrine Tumors After Peptide Receptor Radionuclide Therapy: A Systematic Review. JAMA Oncol 2021; 6:1086-1092. [PMID: 32297906 DOI: 10.1001/jamaoncol.2020.0078] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Importance Peptide receptor radionuclide therapy (PRRT) is a tumor-targeted treatment that uses radiation to induce tumor cell death in neuroendocrine tumors (NET) via β particle-emitting radionuclide linked to a somatostatin peptide analog. Therapy-related myeloid neoplasm (t-MN) has been reported as a potential long-term and frequently lethal adverse event after PRRT. However, the incidence, time of diagnosis, and nature of t-MN is unclear. Therefore, a systematic review is helpful to study the incidence and characteristics of t-MN after PRRT in patients with NET. Objective To systematically evaluate the literature and report the incidence, time of diagnosis, and nature of t-MN after PRRT. Evidence Review MEDLINE, Embase, Scopus, Web of Science, and Cochrane Central Register of Controlled Trials for articles and abstracts reporting studies of different designs studying more than 1 patient (randomized clinical trials, prospective phase I or phase II, retrospective studies, and case series) were searched from database inception through April 2019. Studies of interest included patients with NET who were treated with PRRT and reported the incidence of t-MN, if any. The primary outcome was the incidence of t-MN. Findings Twenty-eight articles were identified comprising 7334 patients who were treated with PRRT for NET. The main reason of exclusion was not reporting the t-MN incidence. The incidence of t-MN was variable between studies with mean (SD) incidence of 2.61% (4.38%). Of all 134 cases, cytogenetic abnormalities were reported in 32 patients with the most common abnormality being complex cytogenetics, consistent with myeloid neoplasms following exposure to alkylating agents or irradiation. Conclusions and Relevance The risk of t-MN after PRRT is small but not insignificant given the poor prognosis after t-MN diagnosis. Close monitoring is warranted to identify such patients early in the disease course when hematologic abnormalities persist.
Collapse
Affiliation(s)
| | | | - Talal Hilal
- University of Mississippi Medical Center, Jackson
| |
Collapse
|
25
|
Betella N, Smiroldo V, Baldelli R, Lania A. Treatment of NETs from Rare Origin. NEUROENDOCRINE NEOPLASIA MANAGEMENT 2021:211-229. [DOI: 10.1007/978-3-030-72830-4_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Neuroendocrine Lung Cancer Mouse Models: An Overview. Cancers (Basel) 2020; 13:cancers13010014. [PMID: 33375066 PMCID: PMC7792789 DOI: 10.3390/cancers13010014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Neuroendocrine lung tumors are a heterogeneous group of malignancies that share a common neuroendocrine nature. They range from low- and intermediate-grade typical and atypical carcinoma, to the highly malignant large cell neuroendocrine lung carcinoma and small cell carcinoma, with marked differences in incidences and prognosis. This review delineates the current knowledge of the genetic landscape of the human tumors, its influence in the development of genetically engineered mouse models (GEMMs) and the molecular imaging tools available to detect and monitor these diseases. While small cell lung carcinoma is one of the diseases best represented by GEMMs, there is a worrying lack of animal models for the other members of the group, these being understudied diseases. Regardless of the incidence and material available, they all are in urgent need of effective therapies. Abstract Neuroendocrine lung tumors comprise a range of malignancies that extend from benign tumorlets to the most prevalent and aggressive Small Cell Lung Carcinoma (SCLC). They also include low-grade Typical Carcinoids (TC), intermediate-grade Atypical Carcinoids (AC) and high-grade Large Cell Neuroendocrine Carcinoma (LCNEC). Optimal treatment options have not been adequately established: surgical resection when possible is the choice for AC and TC, and for SCLC chemotherapy and very recently, immune checkpoint inhibitors. Some mouse models have been generated based on the molecular alterations identified in genomic analyses of human tumors. With the exception of SCLC, there is a limited availability of (preclinical) models making their development an unmet need for the understanding of the molecular mechanisms underlying these diseases. For SCLC, these models are crucial for translational research and novel drug testing, given the paucity of human material from surgery. The lack of early detection systems for lung cancer point them out as suitable frameworks for the identification of biomarkers at the initial stages of tumor development and for testing molecular imaging methods based on somatostatin receptors. Here, we review the relevant models reported to date, their impact on the understanding of the biology of the tumor subtypes and their relationships, as well as the effect of the analyses of the genetic landscape of the human tumors and molecular imaging tools in their development.
Collapse
|
27
|
Jaykel TJ, Clark MS, Adamo DA, Welch BT, Thompson SM, Young JR, Ehman EC. Thoracic positron emission tomography: 18F-fluorodeoxyglucose and beyond. J Thorac Dis 2020; 12:6978-6991. [PMID: 33282403 PMCID: PMC7711422 DOI: 10.21037/jtd-2019-cptn-09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ongoing technologic and therapeutic advancements in medicine are now testing the limits of conventional anatomic imaging techniques. The ability to image physiology, rather than simply anatomy, is critical in the management of multiple disease processes, especially in oncology. Nuclear medicine has assumed a leading role in detecting, diagnosing, staging and assessing treatment response of various pathologic entities, and appears well positioned to do so into the future. When combined with computed tomography (CT) or magnetic resonance imaging (MRI), positron emission tomography (PET) has become the sine quo non technique of evaluating most solid tumors especially in the thorax. PET/CT serves as a key imaging modality in the initial evaluation of pulmonary nodules, often obviating the need for more invasive testing. PET/CT is essential to staging and restaging in bronchogenic carcinoma and offers key physiologic information with regard to treatment response. A more recent development, PET/MRI, shows promise in several specific lung cancer applications as well. Additional recent advancements in the field have allowed PET to expand beyond imaging with 18F-flurodeoxyglucose (FDG) alone, now with the ability to specifically image certain types of cell surface receptors. In the thorax this predominantly includes 68Ga-DOTATATE which targets the somatostatin receptors abundantly expressed in neuroendocrine tumors, including bronchial carcinoid. This receptor targeted imaging technique permits targeting these tumors with therapeutic analogues such as 177Lu labeled DOTATATE. Overall, the proper utilization of PET in the thorax has the ability to directly impact and improve patient care.
Collapse
Affiliation(s)
| | - Michael S Clark
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel A Adamo
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brain T Welch
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jason R Young
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric C Ehman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
28
|
Davenport AP, Scully CCG, de Graaf C, Brown AJH, Maguire JJ. Advances in therapeutic peptides targeting G protein-coupled receptors. Nat Rev Drug Discov 2020; 19:389-413. [PMID: 32494050 DOI: 10.1038/s41573-020-0062-z] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Dysregulation of peptide-activated pathways causes a range of diseases, fostering the discovery and clinical development of peptide drugs. Many endogenous peptides activate G protein-coupled receptors (GPCRs) - nearly 50 GPCR peptide drugs have been approved to date, most of them for metabolic disease or oncology, and more than 10 potentially first-in-class peptide therapeutics are in the pipeline. The majority of existing peptide therapeutics are agonists, which reflects the currently dominant strategy of modifying the endogenous peptide sequence of ligands for peptide-binding GPCRs. Increasingly, novel strategies are being employed to develop both agonists and antagonists, to both introduce chemical novelty and improve drug-like properties. Pharmacodynamic improvements are evolving to allow biasing ligands to activate specific downstream signalling pathways, in order to optimize efficacy and reduce side effects. In pharmacokinetics, modifications that increase plasma half-life have been revolutionary. Here, we discuss the current status of the peptide drugs targeting GPCRs, with a focus on evolving strategies to improve pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | | | | | | | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
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: 45] [Impact Index Per Article: 9.0] [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
|
30
|
Haider M, Al-Toubah T, El-Haddad G, Strosberg J. Molecular imaging and radionuclide therapy of neuroendocrine tumors. Curr Opin Endocrinol Diabetes Obes 2020; 27:16-21. [PMID: 31789833 DOI: 10.1097/med.0000000000000519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Neuroendocrine tumors are heterogeneous neoplasms with variable prognoses and clinical behaviors. The majority of well differentiated NETs express somatostatin receptors. Identification of these receptors has contributed to advancements in molecular and targeted radiotherapies. RECENT FINDINGS Molecular scans provide important diagnostic, staging, and prognostic data. Somatostatin-receptor imaging aids in selection of patients who are eligible for somatostatin-receptor-targeting therapies. Peptide receptor radionuclide therapy has recently demonstrated robust efficacy in a phase III study of progressive midgut NETs. Current studies are investigating novel receptor agonists and antagonists, new classes of radioactive isotopes, and radiosensitizing combination treatments. SUMMARY The sophistication of molecular imaging is improving and its importance is increasing as a diagnostic, predictive, and prognostic tool. Theranostics, the coupling of molecular imaging with receptor-targeted therapy, represents a novel approach to cancer treatment.
Collapse
Affiliation(s)
- Mintallah Haider
- Department of GI Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | | | | | | |
Collapse
|
31
|
McGarrah PW, Westin GFM, Hobday TJ, Scales JA, Ingimarsson JP, Leibovich BC, Halfdanarson TR. Renal Neuroendocrine Neoplasms: A Single-center Experience. Clin Genitourin Cancer 2019; 18:e343-e349. [PMID: 31911122 DOI: 10.1016/j.clgc.2019.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Primary neuroendocrine neoplasms (NENs) of the kidney are exceedingly rare malignancies and the available literature is very limited. The natural history and response to treatments is not well characterized. We aimed to describe the presenting features, demographics, tumor characteristics, and treatment outcomes of patients with renal NENs. PATIENTS AND METHODS We performed a retrospective analysis of all Mayo Clinic patient records with a tissue diagnosis of a primary renal NEN. Baseline patient and surgical pathologic features and treatment modalities were collected. Time to recurrence after resection and overall survival (OS) were estimated using with survival analysis. Surveillance, Epidemiology, and End Results data were used to estimate the population-wide incidence and OS. RESULTS A total of 17 patients were included in the present study, with a median follow-up of 62.8 months. Distant metastasis was present in 29% at diagnosis, with 76% experiencing distant metastasis at any point; 24% had a horseshoe kidney. Of the 17 patients, 14 had undergone surgical resection with no evidence of disease postoperatively. Ten of these patients had documented recurrence. The median time to recurrence was 18 months (95% confidence interval, 9-46 months). Only 1 of the 10 patients showed a radiographic response to systemic therapy. Of 9 patients, 4 had stable disease with somatostatin analogs. The median OS was 143 months (95% confidence interval, 50-143 months). CONCLUSIONS Renal NENs are rare malignancies affecting mostly middle-age patients, with distant metastasis being common. Approximately one half of patients experience stable disease with somatostatin analogs. The OS usually exceeds 5 years.
Collapse
Affiliation(s)
| | - Gustavo F M Westin
- Division of Medical Oncology, Mayo Clinic, Rochester, MN; University Cancer and Blood Center, Athens, GA
| | | | - Joseph A Scales
- Department of Urology, Mayo Clinic, Rochester, MN; Urology Clinics of North Texas, Plano, TX
| | - Johann P Ingimarsson
- Department of Urology, Mayo Clinic, Rochester, MN; Maine Medical Partners Urology, South Portland, ME
| | | | | |
Collapse
|