51
|
Andrini E, Marchese PV, De Biase D, Mosconi C, Siepe G, Panzuto F, Ardizzoni A, Campana D, Lamberti G. Large Cell Neuroendocrine Carcinoma of the Lung: Current Understanding and Challenges. J Clin Med 2022; 11:jcm11051461. [PMID: 35268551 PMCID: PMC8911276 DOI: 10.3390/jcm11051461] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Large cell neuroendocrine carcinoma of the lung (LCNEC) is a rare and highly aggressive type of lung cancer, with a complex biology that shares similarities with both small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC). The prognosis of LCNEC is poor, with a median overall survival of 8-12 months. The diagnosis of LCNEC requires the identification of neuroendocrine morphology and the expression of at least one of the neuroendocrine markers (chromogranin A, synaptophysin or CD56). In the last few years, the introduction of next-generation sequencing allowed the identification of molecular subtypes of LCNEC, with prognostic and potential therapeutic implications: one subtype is similar to SCLC (SCLC-like), while the other is similar to NSCLC (NSCLC-like). Because of LCNEC rarity, most evidence comes from small retrospective studies and treatment strategies that are extrapolated from those adopted in patients with SCLC and NSCLC. Nevertheless, limited but promising data about targeted therapies and immune checkpoint inhibitors in patients with LCNEC are emerging. LCNEC clinical management is still controversial and standardized treatment strategies are currently lacking. The aim of this manuscript is to review clinical and molecular data about LCNEC to better understand the optimal management and the potential prognostic and therapeutic implications of molecular subtypes.
Collapse
Affiliation(s)
- Elisa Andrini
- Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi University Hospital, ENETS Center of Excellence, 40138 Bologna, Italy; (E.A.); (P.V.M.); (A.A.); (G.L.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi University Hospital, ENETS Center of Excellence, 40138 Bologna, Italy; (E.A.); (P.V.M.); (A.A.); (G.L.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138 Bologna, Italy
| | - Dario De Biase
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, Viale Ercolani 4/2, 40138 Bologna, Italy;
| | - Cristina Mosconi
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, 40138 Bologna, Italy;
| | - Giambattista Siepe
- Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesco Panzuto
- Digestive Disease Unit, ENETS Center of Excellence of Rome, Sant’Andrea University Hospital, 00189 Rome, Italy;
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Andrea Ardizzoni
- Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi University Hospital, ENETS Center of Excellence, 40138 Bologna, Italy; (E.A.); (P.V.M.); (A.A.); (G.L.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138 Bologna, Italy
| | - Davide Campana
- Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi University Hospital, ENETS Center of Excellence, 40138 Bologna, Italy; (E.A.); (P.V.M.); (A.A.); (G.L.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138 Bologna, Italy
- Correspondence:
| | - Giuseppe Lamberti
- Department of Experimental, Diagnostic and Specialty Medicine, Sant’Orsola-Malpighi University Hospital, ENETS Center of Excellence, 40138 Bologna, Italy; (E.A.); (P.V.M.); (A.A.); (G.L.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via P. Albertoni 15, 40138 Bologna, Italy
| |
Collapse
|
52
|
Peng W, Cao L, Chen L, Lin G, Zhu B, Hu X, Lin Y, Zhang S, Jiang M, Wang J, Li J, Li C, Shao L, Du H, Hou T, Chen Z, Xiang J, Pu X, Li J, Xu F, Loong H, Wu L. Comprehensive Characterization of the Genomic Landscape in Chinese Pulmonary Neuroendocrine Tumors Reveals Prognostic and Therapeutic Markers (CSWOG-1901). Oncologist 2022; 27:e116-e125. [PMID: 35641209 PMCID: PMC8895731 DOI: 10.1093/oncolo/oyab044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/07/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pulmonary neuroendocrine tumors (pNETs) include typical carcinoid (TC), atypical carcinoid (AC), large cell neuroendocrine carcinoma (LCNEC), and small cell lung carcinoma (SCLC). The optimal treatment strategy for each subtype remains elusive, partly due to the lack of comprehensive understanding of their molecular features. We aimed to explore differential genomic signatures in pNET subtypes and identify potential prognostic and therapeutic biomarkers. METHODS We investigated genomic profiles of 57 LCNECs, 49 SCLCs, 18 TCs, and 24 ACs by sequencing tumor tissues with a 520-gene panel and explored the associations between genomic features and prognosis. RESULTS Both LCNEC and SCLC displayed higher mutation rates for TP53, PRKDC, SPTA1, NOTCH1, NOTCH2, and PTPRD than TC and AC. Small cell lung carcinoma harbored more frequent co-alterations in TP53-RB1, alterations in PIK3CA and SOX2, and mutations in HIF-1, VEGF and Notch pathways. Large cell neuroendocrine carcinoma (12.7 mutations/Mb) and SCLC (11.9 mutations/Mb) showed higher tumor mutational burdens than TC (2.4 mutations/Mb) and AC (7.1 mutations/Mb). 26.3% of LCNECs and 20.8% of ACs harbored alterations in classical non-small cell lung cancer driver genes. The presence of alterations in the homologous recombination pathway predicted longer progression-free survival in advanced LCNEC patients with systemic therapy (P = .005) and longer overall survival (OS) in SCLC patients with resection (P = .011). The presence of alterations in VEGF (P = .048) and estrogen (P = .018) signaling pathways both correlated with better OS in patients with resected SCLC. CONCLUSION We performed a comprehensive genomic investigation on 4 pNET subtypes in the Chinese population. Our data revealed distinctive genomic signatures in subtypes and provided new insights into the prognostic and therapeutic stratification of pNETs.
Collapse
Affiliation(s)
- Wenying Peng
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Liming Cao
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People’s Republic of China
| | - Gen Lin
- Department of Thoracic Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Xiaohua Hu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yingcheng Lin
- Cancer Hospital of Shantou University Medical College, Shantou, People’s Republic of China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Meilin Jiang
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Jingyi Wang
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Junjun Li
- Department of Pathology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Chao Li
- Department of Pathology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Lin Shao
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Haiwei Du
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Ting Hou
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Zhiqiu Chen
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Jianxing Xiang
- Burning Rock Biotech, Guangzhou, People’s Republic of China
| | - Xingxiang Pu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Jia Li
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Fang Xu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| | - Herbert Loong
- Department of Clinical Oncology, Deputy Medical Director, Phase 1 Clinical Trials Centre, Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Lin Wu
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
53
|
Rindi G, Mete O, Uccella S, Basturk O, La Rosa S, Brosens LAA, Ezzat S, de Herder WW, Klimstra DS, Papotti M, Asa SL. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr Pathol 2022; 33:115-154. [PMID: 35294740 DOI: 10.1007/s12022-022-09708-2] [Citation(s) in RCA: 370] [Impact Index Per Article: 123.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
In this review, we detail the changes and the relevant features that are applied to neuroendocrine neoplasms (NENs) in the 2022 WHO Classification of Endocrine and Neuroendocrine Tumors. Using a question-and-answer approach, we discuss the consolidation of the nomenclature that distinguishes neuronal paragangliomas from epithelial neoplasms, which are divided into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). The criteria for these distinctions based on differentiation are outlined. NETs are generally (but not always) graded as G1, G2, and G3 based on proliferation, whereas NECs are by definition high grade; the importance of Ki67 as a tool for classification and grading is emphasized. The clinical relevance of proper classification is explained, and the importance of hormonal function is examined, including eutopic and ectopic hormone production. The tools available to pathologists for accurate classification include the conventional biomarkers of neuroendocrine lineage and differentiation, INSM1, synaptophysin, chromogranins, and somatostatin receptors (SSTRs), but also include transcription factors that can identify the site of origin of a metastatic lesion of unknown primary site, as well as hormones, enzymes, and keratins that play a role in functional and structural correlation. The recognition of highly proliferative, well-differentiated NETs has resulted in the need for biomarkers that can distinguish these G3 NETs from NECs, including stains to determine expression of SSTRs and those that can indicate the unique molecular pathogenetic alterations that underlie the distinction, for example, global loss of RB and aberrant p53 in pancreatic NECs compared with loss of ATRX, DAXX, and menin in pancreatic NETs. Other differential diagnoses are discussed with recommendations for biomarkers that can assist in correct classification, including the distinctions between epithelial and non-epithelial NENs that have allowed reclassification of epithelial NETs in the spine, in the duodenum, and in the middle ear; the first two may be composite tumors with neuronal and glial elements, and as this feature is integral to the duodenal lesion, it is now classified as composite gangliocytoma/neuroma and neuroendocrine tumor (CoGNET). The many other aspects of differential diagnosis are detailed with recommendations for biomarkers that can distinguish NENs from non-neuroendocrine lesions that can mimic their morphology. The concepts of mixed neuroendocrine and non-neuroendocrine (MiNEN) and amphicrine tumors are clarified with information about how to approach such lesions in routine practice. Theranostic biomarkers that assist patient management are reviewed. Given the significant proportion of NENs that are associated with germline mutations that predispose to this disease, we explain the role of the pathologist in identifying precursor lesions and applying molecular immunohistochemistry to guide genetic testing.
Collapse
Affiliation(s)
- Guido Rindi
- Department of Life Sciences and Public Health, Section of Anatomic Pathology, Università Cattolica del Sacro Cuore, Rome, Italy.
- Department of Woman and Child Health Sciences and Public Health, Anatomic Pathology Unit, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo A. Gemelli, 8, 00168, Rome, Italy.
- ENETS Center of Excellence, Rome, Italy.
| | - Ozgur Mete
- Department of Pathology, University Health Network, University of Toronto, 200 Elizabeth Street, 11th floor, Toronto, ON, M5G 2C4, Canada.
| | - Silvia Uccella
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefano La Rosa
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Lodewijk A A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Shereen Ezzat
- Department of Medicine, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Wouter W de Herder
- Department of Internal Medicine, Sector of Endocrinology, Erasmus MC Cancer Institute, ENETS Center of Excellence Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - David S Klimstra
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Paige.AI, New York, NY, USA
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Sylvia L Asa
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
54
|
Hermans BCM, Derks JL, Hillen LM, van der Baan I, van den Broek EC, von der Thüsen JH, van Suylen R, Atmodimedjo PN, den Toom TD, Coumans‐Stallinga C, Timens W, Dinjens WNM, Dubbink HJ, Speel EM, Dingemans AC. In-depth molecular analysis of combined and co-primary pulmonary large cell neuroendocrine carcinoma and adenocarcinoma. Int J Cancer 2022; 150:802-815. [PMID: 34674268 PMCID: PMC9298697 DOI: 10.1002/ijc.33853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 01/09/2023]
Abstract
Up to 14% of large cell neuroendocrine carcinomas (LCNECs) are diagnosed in continuity with nonsmall cell lung carcinoma. In addition to these combined lesions, 1% to 7% of lung tumors present as co-primary tumors with multiple synchronous lesions. We evaluated molecular and clinicopathological characteristics of combined and co-primary LCNEC-adenocarcinoma (ADC) tumors. Ten patients with LCNEC-ADC (combined) and five patients with multiple synchronous ipsilateral LCNEC and ADC tumors (co-primary) were included. DNA was isolated from distinct tumor parts, and 65 cancer genes were analyzed by next generation sequencing. Immunohistochemistry was performed including neuroendocrine markers, pRb, Ascl1 and Rest. Pure ADC (N = 37) and LCNEC (N = 17) cases were used for reference. At least 1 shared mutation, indicating tumor clonality, was found in LCNEC- and ADC-parts of 10/10 combined tumors but only in 1/5 co-primary tumors. A range of identical mutations was observed in both parts of combined tumors: 8/10 contained ADC-related (EGFR/KRAS/STK11 and/or KEAP1), 4/10 RB1 and 9/10 TP53 mutations. Loss of pRb IHC was observed in 6/10 LCNEC- and 4/10 ADC-parts. The number and intensity of expression of Ascl1 and neuroendocrine markers increased from pure ADC (low) to combined ADC (intermediate) and combined and pure LCNEC (high). The opposite was true for Rest expression. In conclusion, all combined LCNEC-ADC tumors were clonally related indicating a common origin. A relatively high frequency of pRb inactivation was observed in both LCNEC- and ADC-parts, suggesting an underlying role in LCNEC-ADC development. Furthermore, neuroendocrine differentiation might be modulated by Ascl1(+) and Rest(-) expression.
Collapse
Affiliation(s)
- Bregtje C. M. Hermans
- Department of Pulmonary DiseasesMaastricht University Medical Centre+MaastrichtThe Netherlands,GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Jules L. Derks
- Department of Pulmonary DiseasesMaastricht University Medical Centre+MaastrichtThe Netherlands,GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Lisa M. Hillen
- GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands,Department of PathologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Irene van der Baan
- GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands,Department of PathologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | | | - Jan H. von der Thüsen
- Department of PathologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | | | - Peggy N. Atmodimedjo
- Department of PathologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | - T. Dorine den Toom
- Department of PathologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | - Cecile Coumans‐Stallinga
- GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands,Department of PathologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Wim Timens
- Department of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Winand N. M. Dinjens
- Department of PathologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | - Hendrikus J. Dubbink
- Department of PathologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | - Ernst‐Jan M. Speel
- GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands,Department of PathologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Anne‐Marie C. Dingemans
- Department of Pulmonary DiseasesMaastricht University Medical Centre+MaastrichtThe Netherlands,GROW—School for Oncology & Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands,Department of PulmonologyErasmus MC Cancer Institute, University Medical Center RotterdamRotterdamThe Netherlands
| | | |
Collapse
|
55
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
56
|
Long-term outcomes after video-assisted thoracoscopic surgery in pulmonary large-cell neuroendocrine carcinoma. Surg Oncol 2022; 41:101728. [DOI: 10.1016/j.suronc.2022.101728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 11/19/2022]
|
57
|
Rekhtman N. Lung neuroendocrine neoplasms: recent progress and persistent challenges. Mod Pathol 2022; 35:36-50. [PMID: 34663914 PMCID: PMC8695375 DOI: 10.1038/s41379-021-00943-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
This review summarizes key recent developments relevant to the pathologic diagnosis of lung neuroendocrine neoplasms, including carcinoids, small cell lung carcinoma (SCLC), and large cell neuroendocrine carcinoma (LCNEC). Covered are recent insights into the biological subtypes within each main tumor type, progress in pathological diagnosis and immunohistochemical markers, and persistent challenging areas. Highlighted topics include highly proliferative carcinoids and their distinction from small cell and large cell neuroendocrine carcinomas (NECs), the evolving role of Ki67, the update on the differential diagnosis of NEC to include thoracic SMARCA4-deficient undifferentiated tumors, the recent data on SCLC transcriptional subtypes with the emergence of POU2F3 as a novel marker for the diagnosis of SCLC with low/negative expression of standard neuroendocrine markers, and the update on the diagnosis of LCNEC, particularly in biopsies. There has been remarkable recent progress in the understanding of the genetic and expression-based profiles within each type of lung neuroendocrine neoplasm, and it is hoped that these insights will enable the development of novel diagnostic, prognostic, and predictive biomarkers to aid in the pathologic assessment of these tumors in the future.
Collapse
Affiliation(s)
- Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
58
|
Poté N. Histoséminaire de pathologie onco-thoracique : cas n°4. Ann Pathol 2022; 42:151-155. [DOI: 10.1016/j.annpat.2021.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
|
59
|
Li Y, Shi X, Mao B, Wang L, Wu L, Li J, Jiao S. The genomic mutational landscape and its correlation with TMB, PD-L1 expression and CD8+ T cell infiltration in Chinese Lung Large Cell Neuroendocrine Carcinoma. Lung Cancer 2022; 166:161-169. [DOI: 10.1016/j.lungcan.2022.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/10/2021] [Accepted: 01/06/2022] [Indexed: 01/19/2023]
|
60
|
Surgical Principles in the Management of Lung Neuroendocrine Tumors: Open Questions and Controversial Technical Issues. Curr Treat Options Oncol 2022; 23:1645-1663. [PMID: 36269459 PMCID: PMC9768012 DOI: 10.1007/s11864-022-01026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2022] [Indexed: 01/30/2023]
Abstract
OPINION STATEMENT Primary neuroendocrine tumors (NETs) of the lung represent a heterogeneous group of malignancies arising from the endocrine cells, involving different entities, from well differentiated to highly undifferentiated neoplasms. Because of the predominance of poorly differentiated tumors, advanced disease is observed at diagnosis in more than one third of patients making chemo- or chemoradiotherapy the only possible treatment. Complete surgical resection, as defined as anatomical resection plus systematic lymphadenectomy, becomes a reliable curative option only for that little percentage of patients presenting with stage I (N0) high-grade NETs. On the other hand, complete surgical resection is considered the mainstay treatment for localized low- and intermediate-grade NETs. Therefore, in the era of the mini-invasive surgery, their indolent behavior has suggested that parenchyma-sparing resections could be as adequate as the anatomical ones in terms of oncological outcomes, leading to discuss about the correct extent of resection and about the role of lymphadenectomy when dealing with highly differentiated NETs.
Collapse
|
61
|
Song Z, Zou L. Risk factors, survival analysis, and nomograms for distant metastasis in patients with primary pulmonary large cell neuroendocrine carcinoma: A population-based study. Front Endocrinol (Lausanne) 2022; 13:973091. [PMID: 36329892 PMCID: PMC9623680 DOI: 10.3389/fendo.2022.973091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a rapidly progressive and easily metastatic high-grade lung cancer, with a poor prognosis when distant metastasis (DM) occurs. The aim of our study was to explore risk factors associated with DM in LCNEC patients and to perform survival analysis and to develop a novel nomogram-based predictive model for screening risk populations in clinical practice. METHODS The study cohort was derived from the Surveillance, Epidemiology, and End Results database, from which we selected patients with LCNEC between 2004 to 2015 and formed a diagnostic cohort (n = 959) and a prognostic cohort (n = 272). The risk and prognostic factors of DM were screened by univariate and multivariate analyses using logistic and Cox regressions, respectively. Then, we established diagnostic and prognostic nomograms using the data in the training group and validated the accuracy of the nomograms in the validation group. The diagnostic nomogram was evaluated using receiver operating characteristic curves, decision curve analysis curves, and the GiViTI calibration belt. The prognostic nomogram was evaluated using receiver operating characteristic curves, the concordance index, the calibration curve, and decision curve analysis curves. In addition, high- and low-risk groups were classified according to the prognostic monogram formula, and Kaplan-Meier survival analysis was performed. RESULTS In the diagnostic cohort, LCNEC close to bronchus, with higher tumor size, and with higher N stage indicated higher likelihood of DM. In the prognostic cohort (patients with LCNEC and DM), men with higher N stage, no surgery, and no chemotherapy had poorer overall survival. Patients in the high-risk group had significantly lower median overall survival than the low-risk group. CONCLUSION Two novel established nomograms performed well in predicting DM in patients with LCNEC and in evaluating their prognosis. These nomograms could be used in clinical practice for screening of risk populations and treatment planning.
Collapse
|
62
|
Chen H, Ishihara M, Horita N, Kazahari H, Ochiai R, Tanzawa S, Honda T, Ichikawa Y, Watanabe K, Seki N. Effect of Adjuvant and Palliative Chemotherapy in Large Cell Neuroendocrine Carcinoma of the Lung: A Systematic Review and Meta-Analysis. Cancers (Basel) 2021; 13:cancers13235948. [PMID: 34885057 PMCID: PMC8657002 DOI: 10.3390/cancers13235948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Adjuvant chemotherapy revealed a better outcome than surgery only, but there was no statistical difference in patients with stage IA. The small cell lung cancer regimen (SCLC) was frequently selected in adjuvant chemotherapy. The SCLC regimen showed better survival than the non-SCLC regimen as palliative chemotherapy at the endpoint of the odds ratio of mortality after two years. Abstract Background: Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a rare subset of lung carcinoma with poor overall survival. Methods: A systematic review following a meta-analysis of studies was performed to identify the effect of different selections of chemotherapy in LCNEC. Articles providing overall survival data for adjuvant chemotherapy or palliative chemotherapy for LCNEC were eligible. The odds ratio (OR) of mortality at one or two years after chemotherapy was evaluated. Results: A total of 16 reports were finally included in the quantitative synthesis, involving a total of 5916 LCNEC patients. Adjuvant chemotherapy was administered to 1303 patients, and palliative chemotherapy was administered to 313 patients using either a small cell lung cancer (SCLC) or a non-small cell lung cancer (NSCLC) regimen. The OR for adjuvant chemotherapy was 0.73 (95% confidence interval (CI): 0.59 to 0.89, p = 0.002). The SCLC regimen showed an OR of 0.52 (95% CI: 0.11 to 2.38, p = 0.40) after one year, and 0.32 (95% CI: 0.11 to 0.89, p = 0.03) after two years, compared with the NSCLC regimen. Conclusions: Adjuvant chemotherapy for pulmonary large cell neuroendocrine carcinoma improved the outcome after surgery. The SCLC regimen showed better survival than the NSCLC regimen as palliative chemotherapy.
Collapse
Affiliation(s)
- Hao Chen
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
- Correspondence: (H.C.); (N.S.)
| | - Masashi Ishihara
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Nobuyuki Horita
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama 236-0027, Japan;
| | - Hiroki Kazahari
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Ryusuke Ochiai
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Shigeru Tanzawa
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Takeshi Honda
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Yasuko Ichikawa
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Kiyotaka Watanabe
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
| | - Nobuhiko Seki
- Division of Oncology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.I.); (H.K.); (R.O.); (S.T.); (T.H.); (Y.I.); (K.W.)
- Correspondence: (H.C.); (N.S.)
| |
Collapse
|
63
|
The 2021 WHO Classification of Lung Tumors: Impact of advances since 2015. J Thorac Oncol 2021; 17:362-387. [PMID: 34808341 DOI: 10.1016/j.jtho.2021.11.003] [Citation(s) in RCA: 573] [Impact Index Per Article: 143.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
The 2021 World Health Organisation (WHO) Classification of Thoracic Tumours was published earlier this year, with classification of lung tumors being one of the chapters. The principles remain those of using morphology first, supported by immunohistochemistry and then molecular techniques. In 2015, there was particular emphasis on using immunohistochemistry to make classification more accurate. In 2021, there is greater emphasis throughout the book on advances in molecular pathology across all tumor types. Major features within this edition are 1) broader emphasis on genetic testing than in the 2015 WHO Classification, 2) a chapter entirely dedicated to the classification of small diagnostic samples, 3) continued recommendation to document percentages of histological patterns in invasive non-mucinous adenocarcinomas, with utilization of these features to apply a formal grading system, as well as using only invasive size for T-factor size determination in part lepidic non-mucinous lung adenocarcinomas as recommended by the 8th Edition TNM Classification, 4) recognition of spread through airspaces (STAS) as a histological feature with prognostic significance, 5) moving lymphoepithelial carcinoma to squamous cell carcinomas, 6) update on evolving concepts in lung neuroendocrine neoplasm classification, 7) recognition of bronchiolar adenoma/ciliated muconodular papillary tumor (BA/CMPT) as a new entity within the adenoma subgroup, 8) recognition of thoracic SMARCA4-deficient undifferentiated tumor, and 9) inclusion of essential and desirable diagnostic criteria for each tumor.
Collapse
|
64
|
Ciobanu OA, Martin S, Fica S. Perspectives on the diagnostic, predictive and prognostic markers of neuroendocrine neoplasms (Review). Exp Ther Med 2021; 22:1479. [PMID: 34765020 PMCID: PMC8576627 DOI: 10.3892/etm.2021.10914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroendocrine neoplasms (NENs) are a heterogeneous group of rare tumors with different types of physiology and prognosis. Therefore, prognostic information, including morphological differentiation, grade, tumor stage and primary location, are invaluable and contribute to the formulation of treatment decisions. Biomarkers that are currently used, including chromogranin A (CgA), serotonin and neuron-specific enolase, are singular parameters that cannot be used to accurately predict variables associated with tumor growth, including proliferation, metabolic rate and metastatic potential. In addition, site-specific biomarkers, such as insulin and gastrin, cannot be applied to all types of NENs. The clinical application of broad-spectrum markers, as it is the case for CgA, remains controversial despite being widely used. Due to limitations of the currently available mono-analyte biomarkers, recent studies were conducted to explore novel parameters for NEN diagnosis, prognosis, therapy stratification and evaluation of treatment response. Identification of prognostic factors for predicting NEN outcome is a critical requirement for the planning of adequate clinical management. Advances in ‘liquid’ biopsies and genomic analysis techniques, including microRNA, circulating tumor DNA or circulating tumor cells and sophisticated biomathematical analysis techniques, such as NETest or molecular image-based biomarkers, are currently under investigation as potentially novel tools for the management of NENs in the future. Despite these recent findings yielding promising observations, further research is necessary. The present review therefore summarizes the existing knowledge and recent advancements in the exploration of biochemical markers for NENs, with focus on gastroenteropancreatic-neuroendocrine tumors.
Collapse
Affiliation(s)
- Oana Alexandra Ciobanu
- Department of Endocrinology and Diabetes, Elias Hospital, 011461 Bucharest, Romania.,Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, 20021 Bucharest, Romania
| | - Sorina Martin
- Department of Endocrinology and Diabetes, Elias Hospital, 011461 Bucharest, Romania.,Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, 20021 Bucharest, Romania
| | - Simona Fica
- Department of Endocrinology and Diabetes, Elias Hospital, 011461 Bucharest, Romania.,Department of Endocrinology, Carol Davila University of Medicine and Pharmacy, 20021 Bucharest, Romania
| |
Collapse
|
65
|
Vrontis K, Economidou SC, Fotopoulos G. Platinum Doublet plus Atezolizumab as First-line Treatment in Metastatic Large Cell Neuroendocrine Carcinoma: A Single Institution Experience. Cancer Invest 2021; 40:124-131. [PMID: 34601985 DOI: 10.1080/07357907.2021.1988962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Large Cell Neuroendocrine Carcinoma of the Lung (L-LCNEC) is a rare type of neuroendocrine lung cancer that is increasingly diagnosed. However, the optimal management regarding the advanced stage is unclear. The purpose of this article is to present and compare our experience when L-LCNEC is treated as Small Cell Lung Cancer (SCLC). PATIENTS AND METHODS Overall, eight cases of L-LCNEC were included. We retrospectively reviewed medical files and reports by accessing the Institution's Data of patients diagnosed with L-LCNEC from April 2019 until December 2020 and evaluated their response to the combination of Platinum - Etoposide - Atezolizumab as first-line chemotherapy. RESULTS The overall observed response rate (ORR) of 75%. The median PFS was 6.85 months. The median response duration was 5.5 months. CONCLUSIONS Comparing our findings with other retrospective and prospective studies, it seems that the systematic treatment of choice and management in L-LCNEC of the lung should be that of a small cell carcinoma of the lung.
Collapse
|
66
|
Agar C, Geier M, Léveiller G, Lamy R, Bizec JL, Tiercin M, Bernier C, Robinet G, Léna H, Ricordel C, Corre R. Brief Report on the Efficacy of Nivolumab in Patients With Previously Treated Advanced Large-Cell Neuroendocrine Cancer of the Lung. JTO Clin Res Rep 2021; 2:100129. [PMID: 34590004 PMCID: PMC8474479 DOI: 10.1016/j.jtocrr.2020.100129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/19/2020] [Accepted: 12/04/2020] [Indexed: 11/08/2022] Open
Abstract
Introduction The optimal management of large cell neuroendocrine cancer of the lung (LCNEC) is unclear, and data regarding anti–programmed cell death protein 1 (PD-1) antibodies are scarce. This study reports the clinical efficacy of a PD-1 inhibitor in patients with advanced LCNEC. Methods All patients with stage III to IV LCNEC treated with at least one previous cycle of chemotherapy between January 1, 2015 and December 31, 2018 were reviewed retrospectively. Patients were divided into two groups depending on their exposure to nivolumab as second-line treatment or beyond. The primary objective was to assess nivolumab’s efficacy. Results A total of 51 patients with advanced LCNEC from eight centers were analyzed, including 17 who received nivolumab. The PD-1 inhibitor was used as second-line treatment in 77% of cases, with a median number of eight doses (range: 1–62). After nivolumab treatment, the median overall survival was 12.1 months (95% confidence interval [CI]: 7.10–14.20). The objective response rate was 29.4% (95% CI: 10.3–56.0), and median progression-free survival was 3.9 months (95% CI: 1.68–7.17). The programmed death-ligand 1 status was unknown. There was no difference in the efficacy of first-line chemotherapy; the objective response rate was 23.5% (n = four of 17) in the nivolumab group versus 32.4% (n = 11 of 34) in the conventional treatment group, and progression-free survival was 3.5 months (95% CI: 1.7–4.4) versus 2.1 months (95% CI: 1.4–4.2), respectively. Conclusions In a real-world setting, nivolumab seems to be an effective second-line treatment in patients with advanced LCNEC. Large prospective studies in this setting are still required.
Collapse
Affiliation(s)
- Camille Agar
- Centre Hospitalier Universitaire de Rennes, Service de Pneumologie, Université de Rennes 1, Rennes, France
| | - Margaux Geier
- Centre Hospitalier Régional Universitaire Morvan, Service d'Oncologie, Université de Bretagne Occidentale, Brest, France
| | - Guillaume Léveiller
- Centre Hospitalier Yves le Foll, Service de Pneumologie, Saint-Brieuc, France
| | - Régine Lamy
- Centre Hospitalier Bretagne Sud, Service d'Oncologie, Lorient, France
| | - Jean-Louis Bizec
- Centre Hospitalier Bretagne-Atlantique, Service de Pneumologie, Vannes, France
| | - Marie Tiercin
- Centre Hospitalier Saint Malo, Service de Pneumologie, Saint Malo, France
| | - Cyril Bernier
- Centre Hospitalier Rene Pleven, Service de Pneumologie, Dinan, France
| | - Gilles Robinet
- Centre Hospitalier Régional Universitaire Morvan, Service d'Oncologie, Université de Bretagne Occidentale, Brest, France
| | - Hervé Léna
- Centre Hospitalier Universitaire de Rennes, Service de Pneumologie, Université de Rennes 1, Rennes, France.,Institut National de la Santé et de la Recherche Médicale U1242, Chemistry Oncogenesis Stress and Signalling, Centres de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Charles Ricordel
- Centre Hospitalier Universitaire de Rennes, Service de Pneumologie, Université de Rennes 1, Rennes, France.,Institut National de la Santé et de la Recherche Médicale U1242, Chemistry Oncogenesis Stress and Signalling, Centres de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Romain Corre
- Centre Hospitalier Intercommunal de Cornouaille, Service de Pneumologie, Quimper, France
| |
Collapse
|
67
|
Prieto TG, Baldavira CM, Machado-Rugolo J, Farhat C, Olivieri EHR, de Sá VK, da Silva ECA, Balancin ML, Ab Saber AM, Takagaki TY, Cordeiro de Lima VC, Capelozzi VL. Pulmonary Neuroendocrine Neoplasms Overexpressing Epithelial-Mesenchymal Transition Mechanical Barriers Genes Lack Immune-Suppressive Response and Present an Increased Risk of Metastasis. Front Oncol 2021; 11:645623. [PMID: 34527572 PMCID: PMC8435885 DOI: 10.3389/fonc.2021.645623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
Typical carcinoids (TC), atypical carcinoids (AC), large cell neuroendocrine carcinomas (LCNEC), and small cell lung carcinomas (SCLC) encompass a bimodal spectrum of metastatic tumors with morphological, histological and histogenesis differences, The hierarchical structure reveals high cohesiveness between neoplastic cells by mechanical desmosomes barrier assembly in carcinoid tumors and LCNEC, while SCLC does not present an organoid arrangement in morphology, the neoplastic cells are less cohesive. However, the molecular mechanisms that lead to PNENs metastasis remain largely unknown and require further study. In this work, epithelial to mesenchymal transition (EMT) transcription factors were evaluated using a set of twenty-four patients with surgically resected PNENs, including carcinomas. Twelve EMT transcription factors (BMP1, BMP7, CALD1, CDH1, COL3A1, COL5A2, EGFR, ERBB3, PLEK2, SNAI2, STEAP1, and TCF4) proved to be highly expressed among carcinomas and downregulated in carcinoid tumors, whereas upregulation of BMP1, CDH2, KRT14 and downregulation of CAV2, DSC2, IL1RN occurred in both histological subtypes. These EMT transcription factors identified were involved in proliferative signals, epithelium desmosomes assembly, and cell motility sequential steps that support PNENs invasion and metastasis in localized surgically resected primary tumor. We used a two-stage design where we first examined the candidate EMT transcription factors using a whole-genome screen, and subsequently, confirmed EMT-like changes by transmission electron microscopy and then, the EMT-related genes that were differentially expressed among PNENs subtypes were predicted through a Metascape analysis by in silico approach. A high expression of these EMT transcription factors was significantly associated with lymph node and distant metastasis. The sequential steps for invasion and metastasis were completed by an inverse association between functional barrier created by PD-L1 immunosuppressive molecule and EMT transcriptional factors. Our study implicates upregulation of EMT transcription factors to high proliferation rates, mechanical molecular barriers disassembly and increased cancer cell motility, as a critical molecular event leading to metastasis risk in PNENs thus emerging as a promising tool to select and customize therapy.
Collapse
Affiliation(s)
| | | | - Juliana Machado-Rugolo
- Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil.,Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu, Brazil
| | - Cecília Farhat
- Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | | | - Vanessa Karen de Sá
- International Center of Research/CIPE, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Marcelo Luiz Balancin
- Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | | | - Teresa Yae Takagaki
- Division of Pneumology, Instituto do Coração (Incor), Medical School of University of São Paulo, São Paulo, Brazil
| | - Vladmir Cláudio Cordeiro de Lima
- Oncology, Rede D'Or São Paulo, São Paulo, Brazil.,Department of Clinical Oncology, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| |
Collapse
|
68
|
Guo J, Hou L, Zhang W, Dong Z, Zhang L, Wu C. Improving differential diagnosis of pulmonary large cell neuroendocrine carcinoma and small cell lung cancer via a transcriptomic, biological pathway-based machine learning model. Transl Oncol 2021; 14:101222. [PMID: 34530194 PMCID: PMC8450252 DOI: 10.1016/j.tranon.2021.101222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
A transcriptomic, biological pathway-based machine learning model was constructed. This classification model can predict LCNEC from borderline samples. The model may help clinicians choose appropriate therapy for pulmonary NETs patients.
Background Accurately differentiating between pulmonary large cell neuroendocrine carcinomas (LCNEC) and small cell lung cancer (SCLC) is crucial to make appropriate therapeutic decisions. Here, a classifier was constructed based on transcriptome data to improve the diagnostic accuracy for LCNEC and SCLC. Methods 13,959 genes mapped to 186 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were included. Gene Set Variation Analysis (GSVA) algorithm was used to enrich and score each KEGG pathway from RNA-sequencing data of each sample. A prediction model based on GSVA score was constructed and trained via ridge regression based on RNA-sequencing datasets from 3 published studies. It was validated by another independent RNA-sequencing dataset. Clinical feasibility was tested by comparing model predicated result using RNA-sequencing data derived from hard-to-diagnose samples of lung neuroendocrine cancer to conventional histology-based diagnosis. Results This model achieved a ROC-AUC of 0.949 and a concordance rate of 0.75 for the entire prediction efficiency. Of the 27 borderline samples, 17/27 (63.0%) were predicted as LCNEC, 7/27 were predicted as SCLC, and the remainder was NSCLC. Only 8 cases (29.6%) with LCNEC were diagnosed by pathologists, which was significantly lower than the results predicted by the model. Furthermore, cases with predicted LCNEC by the model had a significant longer disease-free survival than those where the model predicted SCLC (P = 0.0043). Conclusion This model was able to give an accurate prediction of LCNEC and SCLC. It may assist clinicians to make the optimal decision for patients with pulmonary neuroendocrine tumors in choosing appropriate treatment.
Collapse
Affiliation(s)
- Junhong Guo
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Wei Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Zhengwei Dong
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, PR China.
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, PR China.
| |
Collapse
|
69
|
Large cell neuroendocrine lung carcinoma: consensus statement from The British Thoracic Oncology Group and the Association of Pulmonary Pathologists. Br J Cancer 2021; 125:1210-1216. [PMID: 34489586 PMCID: PMC8548341 DOI: 10.1038/s41416-021-01407-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/01/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022] Open
Abstract
Over the past 10 years, lung cancer clinical and translational research has been characterised by exponential progress, exemplified by the introduction of molecularly targeted therapies, immunotherapy and chemo-immunotherapy combinations to stage III and IV non-small cell lung cancer. Along with squamous and small cell lung cancers, large cell neuroendocrine carcinoma (LCNEC) now represents an area of unmet need, particularly hampered by the lack of an encompassing pathological definition that can facilitate real-world and clinical trial progress. The steps we have proposed in this article represent an iterative and rational path forward towards clinical breakthroughs that can be modelled on success in other lung cancer pathologies.
Collapse
|
70
|
Prieto TG, Machado-Rugolo J, Baldavira CM, Velosa APP, Teodoro WR, Saber AMA, Capelozzi VL. The Fibrosis-Targeted Collagen/Integrins Gene Profile Predicts Risk of Metastasis in Pulmonary Neuroendocrine Neoplasms. Front Oncol 2021; 11:706141. [PMID: 34458147 PMCID: PMC8385766 DOI: 10.3389/fonc.2021.706141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022] Open
Abstract
Recently, collagen/integrin genes have shown promise as predictors of metastasis mainly in non-small cell lung cancer and breast cancer. However, it is unknown if these gene expression profiling differ in metastatic potential of pulmonary neuroendocrine neoplasms (PNENs). In this study, we sought to identify differentially expressed collagen/integrin genes in PNENs in order to understand the molecular mechanisms underlying the development of stroma-associated fibrosis for invasion and metastasis. We compared collagen/integrin gene expression profiling between PNE tumors (PNETs) and PNE carcinomas (PNECs) using a two-stage design. First, we used PCR Array System for 84 ECM-related genes, and among them, we found COL1A2, COL3A1, COL5A2, ITGA5, ITGAV, and ITGB1 functionally involved in the formation of the stroma-associated fibrosis among PNENs histological subtypes. Second, we examined the clinical association between the six collagen/integrin genes in tumor tissues from 24 patients with surgically excised PNENs. However, the pathological exam of their resected tissues demonstrated that 10 developed lymph node metastasis and 7 distant metastasis. We demonstrated and validated up regulation of the six fibrogenic genes in PNECs and down regulation in PNETs that were significantly associated with metastasis-free and overall survival (P<0.05). Our study implicates up regulation of fibrogenic genes as a critical molecular event leading to lymph node and distant metastasis in PNENs.
Collapse
Affiliation(s)
- Tabatha Gutierrez Prieto
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Juliana Machado-Rugolo
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil.,Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu, Brazil
| | - Camila Machado Baldavira
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Clinical Hospital, University of São Paulo Medical School (USP), Sao Paulo, Brazil
| | - Walcy Rosolia Teodoro
- Rheumatology Division of the Clinical Hospital, University of São Paulo Medical School (USP), Sao Paulo, Brazil
| | - Alexandre Muxfeldt Ab Saber
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Laboratory of Genomics and Histomorphometry, Department of Pathology, University of São Paulo Medical School (USP), São Paulo, Brazil
| |
Collapse
|
71
|
Corbett V, Arnold S, Anthony L, Chauhan A. Management of Large Cell Neuroendocrine Carcinoma. Front Oncol 2021; 11:653162. [PMID: 34513663 PMCID: PMC8432609 DOI: 10.3389/fonc.2021.653162] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Large cell neuroendocrine carcinoma (LCNEC) is a rare, aggressive cancer with a dismal prognosis. The majority of cases occur in the lung and the gastrointestinal tract; however, it can occur throughout the body. Recently advances in the understanding of the molecular underpinnings of this disease have paved the way for additional novel promising therapies. This review will discuss the current best evidence for management of LCNEC and new directions in the classification and treatment of this rare disease. METHODS We performed a PubMed search for "Large cell neuroendocrine carcinoma" and "High grade neuroendocrine carcinoma." All titles were screened for relevance to the management of LCNEC. Papers were included based on relevance to the management of LCNEC. RESULTS Papers were included reviewing both pulmonary and extra pulmonary LCNEC. We summarized the data driven best practices for the management of both early and advanced stage LCNEC. We describe emerging therapies with promising potential. DISCUSSION LCNEC are rare and aggressive neoplasms. In advanced disease, the historical regimen of platinum based therapy in combination with etoposide or irinotecan remains among the commonly used first line therapies, however for extra thoracic LCNEC regimens like FOLFOX, FOLFOIRI and CAPTEM can also be used. Further effective and safe treatment options are desperately needed. Recently, new advances including a new understanding of the genetic subcategories of LCNEC and immunotherapy agents may guide further treatments.
Collapse
Affiliation(s)
- Virginia Corbett
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Susanne Arnold
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Lowell Anthony
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Aman Chauhan
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
72
|
Damen PJJ, Bulthuis VJ, Hanssens PEJ, Lie ST, Fleischeuer R, Melotte V, Wouters KA, Ruland A, Beckervordersandforth J, Speel EJM. WHO grade I meningiomas that show regrowth after gamma knife radiosurgery often show 1p36 loss. Sci Rep 2021; 11:16432. [PMID: 34385566 PMCID: PMC8361078 DOI: 10.1038/s41598-021-95956-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
WHO grade I meningiomas occasionally show regrowth after radiosurgical treatment, which cannot be predicted by clinical features. There is increasing evidence that certain biomarkers are associated with regrowth of meningiomas. The aim of this retrospective study was to asses if these biomarkers could be of value to predict regrowth of WHO grade I meningiomas after additive radiosurgery. Forty-four patients with WHO grade I meningiomas who underwent additive radiosurgical treatment between 2002 and 2015 after Simpson IV resection were included in this study, of which 8 showed regrowth. Median follow-up time was 64 months (range 24–137 months). Tumors were analyzed for the proliferation marker Ki-67 by immunohistochemistry and for deletion of 1p36 by fluorescence in situ hybridization (FISH). Furthermore, genomic DNA was analyzed for promoter hypermethylation of the genes NDRG1–4, SFRP1, HOXA9 and MGMT. Comparison of meningiomas with and without regrowth after radiosurgery revealed that loss of 1p36 (p = 0.001) and hypermethylation of NDRG1 (p = 0.046) were correlated with regrowth free survival. Loss of 1p36 was the only parameter that was significantly associated with meningioma regrowth after multivariate analysis (p = 0.01). Assessment of 1p36 loss in tumor tissue prior to radiosurgery might be considered an indicator of prognosis/regrowth. However, this finding has to be validated in an independent larger set of tumors.
Collapse
Affiliation(s)
- Pim J J Damen
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Vincent J Bulthuis
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Suan Te Lie
- Gamma Knife Center Tilburg, ETZ-Elisabeth Hospital, Tilburg, The Netherlands
| | - Ruth Fleischeuer
- Department of Pathology, ETZ-Elisabeth Hospital, Tilburg, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Kim A Wouters
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Andrea Ruland
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Jan Beckervordersandforth
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands
| | - Ernst Jan M Speel
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, Postbox 5800, 6202 AZ, Maastricht, The Netherlands.
| |
Collapse
|
73
|
Zhang J, Yang L, Li J. [Advances in Molecular Biomarker for Pulmonary Large Cell Neuroendocrine Carcinoma]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 23:983-988. [PMID: 33203199 PMCID: PMC7679220 DOI: 10.3779/j.issn.1009-3419.2020.101.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pulmonary large cell neuroendocrine carcinoma (LCNEC) is a pathological subtype of lung neuroendocrine cancer, which accounts for 2.4%-3.1% in surgical specimens of lung cancer. It is characterized by high invasiveness and poor prognosis, and highly correlated with smoking. There are few relevant studies due to the low incidence and small sample size. Therefore, it is relatively difficult to diagnosis and treatment in clinical practice. In this review, we described molecular subtype, diagnostic and prognostic-related markers about large cell neuroendocrine carcinoma of lung based on the recent progress in genomic sequencing and molecular markers, to find the direction for the next research.
.
Collapse
Affiliation(s)
- Jinyao Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
74
|
Fisch D, Bozorgmehr F, Kazdal D, Kuon J, Klotz LV, Shah R, Eichhorn F, Kriegsmann M, Schneider MA, Muley T, Stenzinger A, Bischoff H, Christopoulos P. Comprehensive Dissection of Treatment Patterns and Outcome for Patients With Metastatic Large-Cell Neuroendocrine Lung Carcinoma. Front Oncol 2021; 11:673901. [PMID: 34307143 PMCID: PMC8295750 DOI: 10.3389/fonc.2021.673901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/23/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Large-cell neuroendocrine lung carcinoma (LCNEC) is a rare pulmonary neoplasm with poor prognosis and limited therapeutic options. METHODS We retrospectively analyzed all patients with metastatic LCNEC in the records of a large German academic center since 2010. RESULTS 191 patients were identified with a predominance of male (68%) smokers (92%) and a median age of 65 years. The single most important factor associated with outcome was the type of systemic treatment, with a median overall survival (OS) of 26.4 months in case of immune checkpoint inhibitor administration (n=13), 9.0 months for other patients receiving first-line platinum doublets (n=129), and 4.0 months with non-platinum chemotherapies (n=17, p<0.01). Other patient characteristics independently associated with longer OS were a lower baseline serum LDH (hazard ratio [HR] 0.54, p=0.008) and fewer initial metastatic sites (HR 0.52, p=0.006), while the platinum drug type (cisplatin vs. carboplatin) and cytotoxic partner (etoposide vs. paclitaxel), patients' smoking status and baseline levels of tumor markers (NSE, CYFRA 21-1, CEA) did not matter. 12% (23/191) of patients forewent systemic treatment, mainly due to tumor-related clinical deterioration (n=13), while patient refusal of therapy (n=5) and severe concomitant illness (n=5) were less frequent. The attrition between successive treatment lines was approximately 50% and similar for platinum-based vs. other therapies, but higher in case of a worse initial ECOG status or higher serum LDH (p<0.05). 19% (36/191) of patients had secondary stage IV disease and showed fewer metastatic sites, better ECOG status and longer OS (median 12.6 vs. 8.7 months, p=0.030). Among the 111 deceased patients with palliative systemic treatment and complete follow-up, after exclusion of oligometastatic cases (n=8), administration of local therapies (n=63 or 57%) was associated with a longer OS (HR 0.58, p=0.008), but this association did not persist with multivariable testing. CONCLUSIONS Highly active systemic therapies, especially immunotherapy and platinum doublets, are essential for improved outcome in LCNEC and influence OS stronger than clinical disease parameters, laboratory results and other patient characteristics. The attrition between chemotherapy lines is approximately 50%, similar to other NSCLC. Patients with secondary metastatic disease have a more favorable clinical phenotype and longer survival.
Collapse
Affiliation(s)
- David Fisch
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Farastuk Bozorgmehr
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jonas Kuon
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Laura V. Klotz
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Rajiv Shah
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Florian Eichhorn
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Mark Kriegsmann
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc A. Schneider
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Helge Bischoff
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
75
|
Popper H, Brcic L. Diagnosis and Molecular Profiles of Large Cell Neuroendocrine Carcinoma With Potential Targets for Therapy. Front Oncol 2021; 11:655752. [PMID: 34307132 PMCID: PMC8293294 DOI: 10.3389/fonc.2021.655752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/22/2021] [Indexed: 11/29/2022] Open
Abstract
Large cell neuroendocrine carcinoma (LCNEC) together with small cell carcinoma (SCLC) and typical and atypical carcinoids form the group of pulmonary neuroendocrine tumors. LCNEC and SCLC are high-grade carcinomas. Although both can be found outside the thoracic cavity, they are most common in the lung. LCNEC differs from SCLC by morphologic pattern, and by cytological features such as nuclear size, nucleoli, chromatin pattern, but also by genetic differences. Originally thought to represent a single entity, it became evident, that three subgroups of LCNEC can be identified at the molecular level: a SCLC-like type with loss of retinoblastoma 1 gene (RB1) and TP53 mutations; a non-small cell lung carcinoma (NSCLC)-like type with wildtype RB1, TP53 mutation, and activating mutations of the phosphoinositol-3 kinase (PI3K-CA), or loss of PTEN; and a carcinoid-like type with MEN1 gene mutation. These subtypes can be identified by immunohistochemical staining for RB1, p53, and molecular analysis for PI3K and MEN1 mutations. These subtypes might also respond differently to chemotherapy. Immuno-oncologic treatment has also been applied to LCNEC, however, in addition to the evaluation of tumor cells the stroma evaluation seems to be important. Based on personal experiences with these tumors and available references this review will try to encompass our present knowledge in this rare entity and provoke new studies for better treatment of this carcinoma.
Collapse
Affiliation(s)
- Helmut Popper
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
76
|
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
|
77
|
Atieh T, Huang CH. Treatment of Advanced-Stage Large Cell Neuroendocrine Cancer (LCNEC) of the Lung: A Tale of Two Diseases. Front Oncol 2021; 11:667468. [PMID: 34178652 PMCID: PMC8226095 DOI: 10.3389/fonc.2021.667468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
LCNEC of the lung comprises a small proportion of pulmonary malignancies. Traditionally, they have been classified based on histologic and immunohistochemistry characteristics with features of small cell and non-small cell lung cancer. The treatment outcome of advanced-stage LCNEC of the lung is poor with response rates ranging from 34 to 46% with platinum doublets, median progression-free survival (mPFS) ranging between 4.4 and 5.8 m, and median overall survival (mOS) ranging from 8 to 12.6 m. The optimal treatment strategy for LCNEC is debated given limited data and different outcomes based on chemotherapy type reported in the available literature. Recently, genomic profiling with Next Generation Sequencing (NGS) has been able to sub-classify LCNEC as SCLC-like or NSCLC-like. Treatment based on this sub-classification has improved outcomes by using SCLC and NSCLC regimens based on their genomic profile in retrospective analysis. Future studies in LCNEC of the lung should incorporate this new molecular sub-classification as stratification and possibly include SCLC-like LCNEC into SCLC studies and NSCLC-like into NSCLC studies.
Collapse
Affiliation(s)
- Tahani Atieh
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Chao H Huang
- Division of Medical Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, KS, United States.,Subpecialty Medicine, Kansas City VA Medical Center, Kansas City, MO, United States
| |
Collapse
|
78
|
Hadoux J, Kanaan C, Durand A, Hescot S, Hautefeuille V, Cadiot G, Tauveron I, Laboureau S, Do Cao C, Walter T, Petorin C, Blanchet O, Jannin A, Gu C, Faron M, Leteurtre E, Rousselet MC, Zakeyh JJ, Marchal A, Chatelain D, Beaulaton C, Hervieu V, Ducreux M, Scoazec JY, Baudin E. Prognostic factors of metastatic neuroendocrine carcinoma under first-line treatment with platinum etoposide with a focus on NEC score and Rb expression: Results from the multicentre RBNEC study of the Groupe d'Etude des Tumeurs Endocrines (GTE) and the ENDOCAN-RENATEN network. Eur J Cancer 2021; 152:100-115. [PMID: 34090142 DOI: 10.1016/j.ejca.2021.04.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION AND AIM Neuroendocrine carcinomas (NECs) are aggressive malignant diseases. Platinum-etoposide (PE) combination is the standard first-line treatment, whatever the primary location. The NEC score and also retinoblastoma protein (Rb) status have been suggested to be predictive/prognostic factors in NEC. The primary objective of our multicentric retrospective study was to evaluate the prognostic relevance of the NEC score and Rb status, assessed by immunohistochemistry in PE-treated patients with metastatic NEC. METHODS Seven centres participated. The inclusion criteria were NEC, whatever the primary site, metastatic stage, first-line treatment with PE and tissue samples available. Rb status was determined centrally. RESULTS We report multicentric data from 185 metastatic patients (37% women, median age 63). There were 108 small-cell NECs (SCNECs, 58.4%), 50 large-cell NECs (LCNECs, 27%) and 27 not otherwise specified NECs (nosNECs, 14.6%). The primary sites were the thorax (37%), gastroenteropancreatic sites (38%), unknown (15%) and other (9%). The mean Ki-67 index was 76% (range 20-100). Rb status was interpretable in 122 cases. Rb expression was lost in 74% of the cases: 84% of SCNEC vs. 60% and 63% of LCNEC and nosNEC, respectively (p = 0.016). Objective response was seen in 70% of SCNEC, 45% of LCNEC and 48% of nosNEC (p < 0.001) and in 62% of Rb-negative tumours vs. 46% of Rb-positive tumours (p = 0.3). There was no difference in median progression-free survival or overall survival (OS) as per Rb status. Age, NEC score and response to chemotherapy were the main factors associated with OS in our cohort. CONCLUSION In our series, Rb status had no prognostic impact in PE-treated metastatic patients with NEC, whereas age, NEC score and response to chemotherapy were the main factors associated with OS.
Collapse
Affiliation(s)
- Julien Hadoux
- Oncologie endocrinienne, Département d'imagerie, Gustave Roussy, Villejuif, F-94805, France.
| | - Christina Kanaan
- Service de Pathologie, Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, F-94805, France
| | - Alice Durand
- Service d'oncologie, ENETS Centre of Excellence, Hospices Civils de Lyon et université de Lyon, Lyon, France
| | - Ségolène Hescot
- Département d'oncologie, Institut Curie, Paris, F-75005, France
| | | | - Guillaume Cadiot
- Département d'hépato-gastro-entérologie, CHU de Reims, Reims, France
| | - Igor Tauveron
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHU Clermont-Ferrand, F-63003, Clermont-Ferrand, France; Laboratoire GReD, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Sandrine Laboureau
- Département d'Endocrinologie-Diabétologie-Nutrition, CHU d'Angers, 4 rue Larrey, Angers cedex 9, F-49933, France
| | - Christine Do Cao
- Département d'Endocrinologie, CHU de Lille, Lille, F-59000, France
| | - Thomas Walter
- Service d'oncologie, ENETS Centre of Excellence, Hospices Civils de Lyon et université de Lyon, Lyon, France
| | - Caroline Petorin
- CHU Clermont-Ferrand, Service de Chirurgie Digestive et Hépatobiliaire, 1 rue Lucie et Raymond Aubrac, F-63003, Clermont-Ferrand, France
| | - Odile Blanchet
- CRB, CHU d'Angers, 4 rue Larrey, Angers cedex 9, F-49933, France
| | - Arnaud Jannin
- Département d'Endocrinologie, CHU de Lille, Lille, F-59000, France
| | - Céline Gu
- Service d'anatomo-pathologie, Institut Curie, Paris, F-75005, France
| | - Matthieu Faron
- Département de chirurgie, Gustave Roussy, Villejuif, F-94805, France
| | - Emmanuelle Leteurtre
- CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Université de Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277, Lille, F-59000, France
| | | | - Juliette J Zakeyh
- Laboratoire d'Anatomie Pathologique, CHU Clermont-Ferrand, 1 rue Lucie et Raymond Aubrac, Clermont-Ferrand, F-63003, France
| | - Aude Marchal
- Service d'anatomo-pathologie, CHU Reims, Reims, France
| | | | - Clément Beaulaton
- Service d'anatomo-pathologie, Institut Curie, Paris, F-75005, France
| | - Valérie Hervieu
- Service d'anatomo-pathologie, ENETS Centre of Excellence, Hospices Civils de Lyon et université de Lyon, Lyon, France
| | - Michel Ducreux
- Service d'oncologie digestive, département de médecine, Gustave Roussy, Villejuif, F-94805, France; Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, F-94270, France
| | - Jean-Yves Scoazec
- Service de Pathologie, Département de biologie et pathologie médicale, Gustave Roussy, Villejuif, F-94805, France; Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, F-94270, France
| | - Eric Baudin
- Oncologie endocrinienne, Département d'imagerie, Gustave Roussy, Villejuif, F-94805, France
| |
Collapse
|
79
|
Umakanthan S, Bukelo MM. Concise genetic profile of lung carcinoma. Postgrad Med J 2021; 99:postgradmedj-2021-139860. [PMID: 34083369 DOI: 10.1136/postgradmedj-2021-139860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022]
Abstract
The WHO classification of lung cancer (2015) is based on immunohistochemistry and molecular evaluation. This also includes microscopic analysis of morphological patterns that aids in the pathological diagnosis and classification of lung cancers. Lung cancers are the leading cause of cancer deaths worldwide. Recent advancements in identifying the etiopathogenesis are majorly driven by gene mutation studies. This has been explained by The Cancer Genome Atlas, next-generation sequencer and TRAcking non-small cell lung cancer evolution through therapy [Rx]. This article reviews the genetic profile of adenocarcinoma, squamous cell carcinoma, small cell carcinoma, large cell neuroendocrine carcinoma and pulmonary carcinoids. This includes the prolific genetic alterations and novel molecular changes seen in these tumours. In addition, target- specific drugs that have shown promising effects in clinical use and trials are also briefly discussed.
Collapse
Affiliation(s)
- Srikanth Umakanthan
- Paraclinical Sciences, The University of the West Indies at Saint Augustine Faculty of Medical Sciences, Saint Augustine, Trinidad and Tobago
| | - Maryann M Bukelo
- Department of Anatomical Pathology, Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Mount Hope, Trinidad and Tobago.,Department of Anatomical Patholgy, Laboratory Services, Eric Williams Medical Sciences Complex, North Central Regional Health Authority, Mount Hope, Trinidad and Tobago
| |
Collapse
|
80
|
Saghaeiannejad Esfahani H, Vela CM, Chauhan A. Prevalence of TP-53/Rb-1 Co-Mutation in Large Cell Neuroendocrine Carcinoma. Front Oncol 2021; 11:653153. [PMID: 34141612 PMCID: PMC8203494 DOI: 10.3389/fonc.2021.653153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
Introduction Large cell neuroendocrine carcinoma (LCNEC) is a rare and highly aggressive high-grade neuroendocrine neoplasm, which can arise from anywhere in the body. Due to its rarity there is a lacuna in our understanding of LCNEC's molecular biology. In 2016, Rekhtman and colleagues presented one of the largest molecular sequencing series of pulmonary LCNEC. They differentiated genomic profiles of LCNEC into two major subsets: small cell lung cancer (SCLC)-like, characterized by TP53 + RB1 co-mutation/loss, and non-small cell lung cancer (NSCLC)-like, characterized by the lack of co-altered TP53 + RB1. This finding is of significance because at present LCNEC patients are often treated like SCLC. However, the universal genomic SCLC biomarker of TP53 and RB1 co-mutation was only found in 40% of their cohort. Since then various other scientists have looked into molecular profiling of LCNEC with markedly discordant results. The objective of this study was to conduct a systematic review of publicly available next generation sequencing (NGS) data to evaluate the prevalence of TP53 + RB1 co-mutation in LCNEC. Method We conducted a literature search using PubMed. Seven studies including 302 patients with pulmonary LCNEC and four studies including 20 patients with extra-pulmonary LCNEC underwent final analysis. Results The prevalence of TP53 + RB1 co-mutation was 36% (109/302) among pulmonary LCNEC patients and 35% (7/20) among the extra-thoracic LCNEC cohort. This finding is in stark contrast to >90% TP53 + RB1 co-mutation in SCLC. Conclusion It is now well established that LCNEC is molecularly distinct from SCLC. LCNEC seems to have two molecularly defined sub-cohort based on TP53 + RB1 co-mutation status. Future studies should look into prognostic and predictive implication of TP53 + RB1 co-mutation status in LCNEC. Prospective studies should be designed to characterize molecular subtypes and direct treatment accordingly. We are currently conducting a prospective pilot clinical trial wherein LCNEC patients are treated based on TP53 + RB1 co-mutation status. The study is currently enrolling. "Next Generation Sequencing-Based Stratification of Front Line Treatment of Neuroendocrine Carcinoma (PRECISION-NEC). Systematic Review ClinicalTrials.gov, identifier NCT04452292.
Collapse
Affiliation(s)
| | - Cory M Vela
- Moffitt Cancer Center, University of South Florida, Tampa, FL, United States
| | - Aman Chauhan
- College of Medicine, University of Kentucky, Lexington, KY, United States.,Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
81
|
Abdel-Rahman O, Fazio N. Outcomes of small-cell versus large-cell gastroenteropancreatic neuroendocrine carcinomas: A population-based study. J Neuroendocrinol 2021; 33:e12971. [PMID: 33870570 DOI: 10.1111/jne.12971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 11/29/2022]
Abstract
The recent World Health Organization classification for gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) classified poorly differentiated GEP-NENs into small cell and large cell categories. The present study aimed to assess the differences in outcomes between patients with both histological categories. The Surveillance, Epidemiology and End Results (SEER) database (1975-2016) was accessed and patients with small cell and large cell GEP-neuroendocrine carcinomas (NECs) were extracted. Differences in survival outcomes were explored through Kaplan-Meier survival estimates and multivariable Cox regression models. In total, 2204 patients with GEP-NEC were identified in the survival cohort, including 1698 patients with small cell NEC (77%) and 506 patients with large cell NEC (23%). Using Kaplan-Meier analysis/log-rank testing, large cell GEP-NEC was associated with better overall survival compared to small cell NEC (P < 0.01). Using multivariable Cox regression analysis, large cell GEP-NEC was associated with better overall survival (large cell GEP-NEC versus small cell GEP-NEC, hazard ratio = 0.77; 95% confidence interval = 0.68-0.86) and cancer-specific survival (large cell GEP-NEC versus small cell GEP-NEC, hazard ratio = 0.79; 95% 95% confidence interval = 0.69-0.91). Patients with small cell GEP-NEC have worse survival outcomes compared to those with large cell GEP-NEC. Further efforts are needed to identify biological differences and treatment sensitivities between both histological categories.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO); IRCCS, Milan, Italy
| |
Collapse
|
82
|
Yoshimura M, Seki K, Bychkov A, Fukuoka J. Molecular Pathology of Pulmonary Large Cell Neuroendocrine Carcinoma: Novel Concepts and Treatments. Front Oncol 2021; 11:671799. [PMID: 33968782 PMCID: PMC8100606 DOI: 10.3389/fonc.2021.671799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023] Open
Abstract
Pulmonary large cell neuroendocrine carcinoma (LCNEC) is an aggressive neoplasm with poor prognosis. Histologic diagnosis of LCNEC is not always straightforward. In particular, it is challenging to distinguish small cell lung carcinoma (SCLC) or poorly differentiated carcinoma from LCNEC. However, histological classification for LCNEC as well as their therapeutic management has not changed much for decades. Recently, genomic and transcriptomic analyses have revealed different molecular subtypes raising hopes for more personalized treatment. Two main molecular subtypes of LCNEC have been identified by studies using next generation sequencing, namely type I with TP53 and STK11/KEAP1 alterations, alternatively called as non-SCLC type, and type II with TP53 and RB1 alterations, alternatively called as SCLC type. However, there is still no easy way to classify LCNEC subtypes at the actual clinical level. In this review, we have discussed histological diagnosis along with the genomic studies and molecular-based treatment for LCNEC.
Collapse
Affiliation(s)
| | - Kurumi Seki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junya Fukuoka
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
83
|
Ferrara MG, Stefani A, Simbolo M, Pilotto S, Martini M, Lococo F, Vita E, Chiappetta M, Cancellieri A, D’Argento E, Trisolini R, Rindi G, Scarpa A, Margaritora S, Milella M, Tortora G, Bria E. Large Cell Neuro-Endocrine Carcinoma of the Lung: Current Treatment Options and Potential Future Opportunities. Front Oncol 2021; 11:650293. [PMID: 33937057 PMCID: PMC8081906 DOI: 10.3389/fonc.2021.650293] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Large-cell neuroendocrine carcinomas of the lung (LCNECs) are rare tumors representing 1-3% of all primary lung cancers. Patients with LCNEC are predominantly male, older, and heavy smokers. Histologically, these tumors are characterized by large cells with abundant cytoplasm, high mitotic rate, and neuroendocrine immunohistochemistry-detected markers (chromogranin-A, synaptophysin, and CD56). In 2015 the World Health Organization classified LCNEC as a distinct subtype of pulmonary large-cell carcinoma and, therefore, as a subtype of non-small cell lung carcinoma (NSCLC). Because of the small-sized tissue samples and the likeness to other neuroendocrine tumors, the histological diagnosis of LCNEC remains difficult. Clinically, the prognosis of metastatic LCNECs is poor, with high rates of recurrence after surgery alone and overall survival of approximately 35% at 5 years, even for patients with early stage disease that is dramatically shorter compared with other NSCLC subtypes. First-line treatment options have been largely discussed but with limited data based on phase II studies with small sample sizes, and there are no second-line well defined treatments. To date, no standard treatment regimen has been developed, and how to treat LCNEC is still on debate. In the immunotherapy and targeted therapy era, in which NSCLC treatment strategies have been radically reshaped, a few data are available regarding these opportunities in LCNEC. Due to lack of knowledge in this field, many efforts have been done for a deeper understanding of the biological and molecular characteristics of LCNEC. Next generation sequencing analyses have identified subtypes of LCNEC that may be relevant for prognosis and response to therapy, but further studies are needed to better define the clinical impact of these results. Moreover, scarce data exist about PD-L1 expression in LCNEC and its predictive value in this histotype with regard to immunotherapy efficacy. In the literature some cases are reported concerning LCNEC metastatic patients carrying driver mutations, especially EGFR alterations, showing targeted therapy efficacy in this setting of disease. Due to the rarity and the challenging understanding of LCNEC, in this review we aim to summarize the management options currently available for treatment of LCNEC.
Collapse
Affiliation(s)
- Miriam Grazia Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Medicine, University of Verona Hospital Trust, Verona, Italy
| | - Maurizio Martini
- Institute of Pathology, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Filippo Lococo
- Thoracic Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Marco Chiappetta
- Thoracic Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandra Cancellieri
- Institute of Pathology, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Ettore D’Argento
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Rocco Trisolini
- Interventional Pulmonology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Guido Rindi
- Institute of Pathology, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy
| | - Stefano Margaritora
- Thoracic Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona Hospital Trust, Verona, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
84
|
Miyagawa-Hayashino A, Okada S, Takeda-Miyata N, Takashima Y, Yamada T, Takemura Y, Uchino J, Inoue M, Takayama K, Konishi E. TTF-1 and c-MYC-defined Phenotypes of Large Cell Neuroendocrine Carcinoma and Delta-like Protein 3 Expression for Treatment Selection. Appl Immunohistochem Mol Morphol 2021; 29:313-320. [PMID: 33031101 PMCID: PMC8132912 DOI: 10.1097/pai.0000000000000875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/01/2020] [Indexed: 12/25/2022]
Abstract
The standard treatment regimen has not yet been established for advanced pulmonary large cell neuroendocrine carcinoma (LCNEC) because of its rarity. LCNEC can be subdivided into 2 mutually exclusive molecular subgroups: STK11/KEAP1 and TP53 mutated with high neuroendocrine expression and transcriptional profile of ASCL1high/DLL3high/NOTCHlow (non-small cell lung carcinoma, NSCLC-like) or RB1 and TP53 mutated with reduced neuroendocrine markers and transcriptional pattern of ASCL1low/DLL3low/NOTCHhigh (small cell lung cancer, SCLC-like). Model-based clustering shows that SCLC has subdivided into 2 major proteomic subsets defined by either TTF-1high/c-MYClow or TTF-1low/c-MYChigh, which may correspond to 2 mutually exclusive molecular subgroups: NSCLC-like or SCLC-like, respectively. We herein investigated whether TTF-1 and c-MYC could be applied to LCNEC to identify distinct subsets immunohistochemically and assessed DLL3 expression in these subsets. The protein expression profile may be useful to select patients for potential efficacy of targeted therapies including aurora kinase inhibitors for MYC alterations or anti-DLL3 antibody-drug conjugates. TTF-1 and c-MYC expression was mutually exclusive in 25 of 27 (93%) cases; TTF-1+/c-MYC- in 10, TTF-1-/c-MYC+ in 15, and TTF-1+/c-MYC+ in 2. DLL3 expression was seen in 15 of 27 cases (56%). All 12 TTF-1+ LCNEC cases were positive for DLL3. Three of 15 (20%) TTF-1-/c-MYC+ cases showed DLL3 positivity. LCNEC could be separated into 2 subsets proteomically defined by TTF-1 and c-MYC expression, which may be suitable to guide treatment selection including aurora kinase inhibitors for c-MYC+ cases. TTF-1 positivity can serve as a surrogate marker for DLL3, but caution is necessary as 20% of TTF-1- cases showed DLL3 positivity.
Collapse
MESH Headings
- Aged
- Carcinoma, Large Cell/metabolism
- Carcinoma, Large Cell/pathology
- Carcinoma, Large Cell/therapy
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Intracellular Signaling Peptides and Proteins/biosynthesis
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Membrane Proteins/biosynthesis
- Middle Aged
- Patient Selection
- Proto-Oncogene Proteins c-myc/metabolism
- Thyroid Nuclear Factor 1/metabolism
Collapse
Affiliation(s)
| | | | | | | | - Tadaaki Yamada
- Pulmonary Medicine, Kyoto Prefectural University of Medicine
| | - Yoshizumi Takemura
- Pulmonary Medicine, Kyoto Prefectural University of Medicine
- Department of Pulmonary Medicine, Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Junji Uchino
- Pulmonary Medicine, Kyoto Prefectural University of Medicine
| | | | - Koichi Takayama
- Pulmonary Medicine, Kyoto Prefectural University of Medicine
| | | |
Collapse
|
85
|
Metovic J, Bianchi F, Rossi G, Barella M, Sonzogni A, Harari S, Papotti M, Pelosi G. Recent advances and current controversies in lung neuroendocrine neoplasms ✰. Semin Diagn Pathol 2021; 38:90-97. [PMID: 33810912 DOI: 10.1053/j.semdp.2021.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 11/11/2022]
Abstract
In the lung, neuroendocrine tumors (NETs), namely typical and atypical carcinoids, and neuroendocrine carcinomas (NECs), grouping small cell carcinoma (SCLC) and large cell neuroendocrine carcinoma (LCNEC), make up for distinct tumor entities according to epidemiological, genetic, pathologic and clinical data. The proper classification is essential in clinical practice for diagnosis, prognosis and therapy purposes. Through an extensive literature survey, three perspectives on lung NENs have been revised: i) criteria and terminology on biopsy or cytology samples of primaries or metastases; ii) carcinoids with elevated mitotic counts and/or Ki-67 proliferation rates; iii) relevance of molecular landscape to identify new tumor entities and therapeutic targets. Furthermore, a dispute about lung NEN development has been raised according to emerging molecular models. We herein provide a pathology update on practical topics in the setting of lung NENs according to the current classification (recent advances). We have also reappraised the development of these tumors by modeling risk factors and natural history of disease (recent controversies). Combining recent advances and controversies may help clarify our biological understanding of lung NENs and give practical information for the clinical decision-making process.
Collapse
Affiliation(s)
- Jasna Metovic
- Department of Oncology, University of Turin, Turin, Italy
| | - Fabrizio Bianchi
- Cancer Biomarker Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giulio Rossi
- Operative Unit of Pathologic Anatomy, Azienda USL Romagna, Hospital Santa Maria delle Croci, Ravenna, Italy
| | - Marco Barella
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Angelica Sonzogni
- Department of Pathology and Laboratory Medicine, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sergio Harari
- Department of Medical Sciences and Community Health, University of Milan, Milan, Italy; Division of Pneumology, San Giuseppe Hospital, IRCCS MultiMedica, Milan, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Inter-Hospital Pathology Division, IRCCS MultiMedica, Milan, Italy.
| |
Collapse
|
86
|
An algorithmic approach utilizing CK7, TTF1, beta-catenin, CDX2, and SSTR2A can help differentiate between gastrointestinal and pulmonary neuroendocrine carcinomas. Virchows Arch 2021; 479:481-491. [PMID: 33733343 DOI: 10.1007/s00428-021-03085-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
Primary gastrointestinal neuroendocrine carcinoma (GI-NEC) cannot be distinguished morphologically from pulmonary neuroendocrine carcinoma (P-NEC). This can present a significant diagnostic challenge in cases where site of origin cannot be readily determined. To identify immunohistochemical (IHC) markers that can be used to reliably distinguish between GI-NECs and P-NECs, we constructed 3-mm tissue microarrays, one containing 13 GI-NECs and one containing 20 P-NECs. IHC was performed on both microarrays using 21 stains: AE1/AE3, CK7, CK20, synaptophysin, chromogranin, CD56, INSM1, SSTR2A, CDX2, SATB2, TTF1, Napsin A, PR, GATA3, PAX8, ISL1, beta-catenin, AFP, SMAD4, Rb, and p53. For GI-NEC, the most strongly expressed marker was synaptophysin (mean H-score 248), while AE1/AE3 was the most strongly expressed in P-NEC (mean H-score 230), which was stronger than in GI-NEC (p = 0.011). Other markers that were stronger overall in P-NEC than in GI-NEC included CK7 (p < 0.0001) and TTF1 (p < 0.0001). Markers that were stronger overall in GI-NEC than in P-NEC included SSTR2A (p = 0.0021), SATB2 (p = 0.018), CDX2 (p = 0.019), and beta-catenin (nuclear; p = 0.029). SMAD4, Rb, and p53 showed similar rates of abnormal protein expression. Based on these results, a stepwise algorithmic approach utilizing CK7, TTF1, beta-catenin, CDX2, and SSTR2A had a 91% overall accuracy in distinguishing these GI-NEC from P-NEC. This was tested on a second cohort of 10 metastatic GI-NEC and 10 metastatic P-NEC, with an accuracy in this cohort of 85% and an overall accuracy of 89% for the 53 cases tested. Our algorithm reasonably discriminates GI-NEC from P-NEC using currently available IHC stains.
Collapse
|
87
|
Derks JL, Speel EJM, Dingemans AMC. Adjuvant Chemotherapy in Patients With Stage I-IIIA Large-Cell Neuroendocrine Carcinoma: Should a Different Approach Be Applied Than for Small-Cell Lung Cancer? J Clin Oncol 2021; 39:1508-1509. [PMID: 33661698 DOI: 10.1200/jco.20.03598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jules L Derks
- Jules L. Derks, MD, PhD, Department of Pulmonary Diseases, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; Ernst-Jan M. Speel, PhD, Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; and Anne-Marie C. Dingemans, MD, PhD, Department of Pulmonology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ernst-Jan M Speel
- Jules L. Derks, MD, PhD, Department of Pulmonary Diseases, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; Ernst-Jan M. Speel, PhD, Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; and Anne-Marie C. Dingemans, MD, PhD, Department of Pulmonology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Anne-Marie C Dingemans
- Jules L. Derks, MD, PhD, Department of Pulmonary Diseases, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; Ernst-Jan M. Speel, PhD, Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands; and Anne-Marie C. Dingemans, MD, PhD, Department of Pulmonology, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
88
|
Jiang M, Zhang X. Antiangiogenesis Combined with Immunotherapy to Treat Advanced Small-Cell Carcinoma of the Esophagus Resistant to Chemotherapy: According to the Guidance of Next-Generation Sequencing. Onco Targets Ther 2021; 14:1613-1621. [PMID: 33688208 PMCID: PMC7936705 DOI: 10.2147/ott.s293733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/13/2021] [Indexed: 12/24/2022] Open
Abstract
A 64-year-old woman admitted to our hospital with the chief complaint of swallowing obstruction was diagnosed as relapsed small-cell carcinoma of the esophagus. Complete remission (CR) was observed after six cycles of irinotecan plus cisplatin therapy. According to the results of a next-generation sequencing analysis of the tumor specimen, anlotinib (12 mg PO q3w) was recommended. After 1 month of anlotinib treatment, the tumor decreased significantly according to computed tomography scan and gastroscopy. However, the disease progressed after 2 months of therapy. A gene analysis of the new puncture sample showed microsatellite instability and a high tumor mutation burden. Immunohistochemistry indicated positive programmed death ligand-1 expression (>1%). Because of these results, the patient was treated with anlotinib (12 mg PO q3w) in combination with toripalimab (240 mg IV drip q3w). After 3 months of therapy, CR was achieved, although progression-free survival had not been reached at the time of publication.
Collapse
Affiliation(s)
- Man Jiang
- Cancer Precision Medical Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, People's Republic of China
| | - Xiaochun Zhang
- Cancer Precision Medical Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, People's Republic of China
| |
Collapse
|
89
|
Uccella S, La Rosa S, Metovic J, Marchiori D, Scoazec JY, Volante M, Mete O, Papotti M. Genomics of High-Grade Neuroendocrine Neoplasms: Well-Differentiated Neuroendocrine Tumor with High-Grade Features (G3 NET) and Neuroendocrine Carcinomas (NEC) of Various Anatomic Sites. Endocr Pathol 2021; 32:192-210. [PMID: 33433884 DOI: 10.1007/s12022-020-09660-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 02/06/2023]
Abstract
High-grade neuroendocrine neoplasms (HG-NENs) are clinically aggressive diseases, the classification of which has recently been redefined. They now include both poorly differentiated NENs (neuroendocrine carcinoma, NECs) and high proliferating well-differentiated NENs (called grade 3 neuroendocrine tumors, G3 NETs, in the digestive system). In the last decade, the "molecular revolution" that has affected all fields of medical oncology has also shed light in the understanding of HG NENs heterogeneity and has provided new diagnostic and therapeutic tools, useful in the management of these malignancies. Considering the kaleidoscopic aspects of HG NENs in various anatomical sites, this review systematically addresses the genomic landscape of such neoplasm throughout the more common thoracic and digestive locations, as well as it will consider other rare but not exceptional primary sites, including the skin, the head and neck, and the urogenital system. The revision of the available literature will then be oriented to understand the translational relevance of molecular data, by analyzing conceptual issues, clinicopathological correlations, and unmet needs in this field.
Collapse
Affiliation(s)
- Silvia Uccella
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Stefano La Rosa
- Institute of Pathology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jasna Metovic
- Department of Oncology, University of Turin, Torino, Italy
| | - Deborah Marchiori
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Jean-Yves Scoazec
- Department of Pathology, Gustave Roussy Cancer Campus, Paris, France
| | - Marco Volante
- Department of Oncology, University of Turin, Torino, Italy
| | - Ozgur Mete
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mauro Papotti
- Department of Oncology, University of Turin, Torino, Italy
| |
Collapse
|
90
|
Chen Y, Zhang J, Huang C, Tian Z, Zhou X, Guo C, Liu H, Li S. Survival outcomes of surgery in patients with pulmonary large-cell neuroendocrine carcinoma: a retrospective single-institution analysis and literature review. Orphanet J Rare Dis 2021; 16:82. [PMID: 33579331 PMCID: PMC7881654 DOI: 10.1186/s13023-021-01730-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pulmonary large-cell neuroendocrine carcinoma (pLCNEC) is a very rare malignancy originating from the lung and bronchus, and its biological behaviour, clinical diagnosis, treatment and prognosis are poorly understood. Thus, the clinical characteristics and surgical treatment-related prognostic factors of this rare disorder must be explored. Results The clinical data of 59 patients (48 males and 11 females) who were treated by surgery and diagnosed with pLCNEC by postoperative pathology at Peking Union Medical College Hospital from April 2004 to April 2019 were analysed retrospectively. The median patient age was 62 years (38–79 years), and the median duration of disease was 2 months (0.5–18 months). Compared with other lung malignancies, pLCNEC lacks specific clinical symptoms and imaging features, and preoperative biopsy pathology is often insufficient to confirm the diagnosis. The corresponding numbers of patients who were classified into stages I, II, III and IV according to the postoperative pathological tumour-nodal-metastasis stage were 25, 12, 15 and 7, respectively. The median overall survival was 36 months (0.9–61.1 months). The 1-year, 3-year and 5-year survival rates were 76.3%, 49% and 44.7%, respectively. The tumour stage exerted a significant effect on survival (Cox multivariate analysis p < 0.05). Conclusions For patients with resectable pLCNEC, multidisciplinary therapy based on surgery may have good survival benefits, and tumour stage is an independent risk factor for the prognosis of pLCNEC.
Collapse
Affiliation(s)
- Yeye Chen
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jiaqi Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Zhenhuan Tian
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xiaoyun Zhou
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Hongsheng Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
91
|
Liu F, Li Y, Ying D, Qiu S, He Y, Li M, Liu Y, Zhang Y, Zhu Q, Hu Y, Liu L, Li G, Pan W, Jin W, Mu J, Cao Y, Liu Y. Whole-exome mutational landscape of neuroendocrine carcinomas of the gallbladder. Signal Transduct Target Ther 2021; 6:55. [PMID: 33563892 PMCID: PMC7873252 DOI: 10.1038/s41392-020-00412-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 12/23/2022] Open
Abstract
Neuroendocrine carcinoma (NEC) of the gallbladder (GB-NEC) is a rare but extremely malignant subtype of gallbladder cancer (GBC). The genetic and molecular signatures of GB-NEC are poorly understood; thus, molecular targeting is currently unavailable. In the present study, we applied whole-exome sequencing (WES) technology to detect gene mutations and predicted somatic single-nucleotide variants (SNVs) in 15 cases of GB-NEC and 22 cases of general GBC. In 15 GB-NECs, the C > T mutation was predominant among the 6 types of SNVs. TP53 showed the highest mutation frequency (73%, 11/15). Compared with neuroendocrine carcinomas of other organs, significantly mutated genes (SMGs) in GB-NECs were more similar to those in pulmonary large-cell neuroendocrine carcinomas (LCNECs), with driver roles for TP53 and RB1. In the COSMIC database of cancer-related genes, 211 genes were mutated. Strikingly, RB1 (4/15, 27%) and NAB2 (3/15, 20%) mutations were found specifically in GB-NECs; in contrast, mutations in 29 genes, including ERBB2 and ERBB3, were identified exclusively in GBC. Mutations in RB1 and NAB2 were significantly related to downregulation of the RB1 and NAB2 proteins, respectively, according to immunohistochemical (IHC) data (p values = 0.0453 and 0.0303). Clinically actionable genes indicated 23 mutated genes, including ALK, BRCA1, and BRCA2. In addition, potential somatic SNVs predicted by ISOWN and SomVarIUS constituted 6 primary COSMIC mutation signatures (1, 3, 30, 6, 7, and 13) in GB-NEC. Genes carrying somatic SNVs were enriched mainly in oncogenic signaling pathways involving the Notch, WNT, Hippo, and RTK-RAS pathways. In summary, we have systematically identified the mutation landscape of GB-NEC, and these findings may provide mechanistic insights into the specific pathogenesis of this deadly disease.
Collapse
Affiliation(s)
- Fatao Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Yongsheng Li
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongjian Ying
- Department of Minimal Invasive Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, 315040, China
| | - Shimei Qiu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Yong He
- Department of hepatopancreatobiliary surgery, Ganzhou hospital affiliated to Nanchang university, Jiangxi, 341000, China
| | - Maolan Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Yun Liu
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yijian Zhang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Qin Zhu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Yunping Hu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
| | - Liguo Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoqiang Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihua Pan
- Department of Pediatric Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
- Information and Big Data Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
| | - Wei Jin
- Information and Big Data Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China
| | - Jiasheng Mu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China.
- Information and Big Data Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, 200092, China.
| | - Yang Cao
- Department of Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
- Department of Gastric Surgery, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| | - Yingbin Liu
- Shanghai Key Laboratory of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China.
- State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
- Shanghai Research Center of Biliary Tract Disease, Yangpu District, Shanghai, 200092, China.
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
92
|
Shirasawa M, Yoshida T, Takayanagi D, Shiraishi K, Yagishita S, Sekine K, Kanda S, Matsumoto Y, Masuda K, Shinno Y, Okuma Y, Goto Y, Horinouchi H, Hamada A, Kohno T, Yamamoto N, Watanabe SI, Ohe Y, Motoi N. Activity and Immune Correlates of Programmed Death-1 Blockade Therapy in Patients With Advanced Large Cell Neuroendocrine Carcinoma. Clin Lung Cancer 2021; 22:282-291.e6. [PMID: 33722498 DOI: 10.1016/j.cllc.2021.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The efficacy of anti-programmed death receptor 1 (PD-1) therapy in patients with large cell neuroendocrine carcinoma (LCNEC) remains unclear. We investigated the outcome of anti-PD-1 therapy and its predictive markers by evaluating the immune-related tumor microenvironment. PATIENTS We retrospectively reviewed patients with advanced LCNEC treated with systemic chemotherapy. We also evaluated PD ligand 1 (PD-L1) expression (clone: 22C3), CD8-positive tumor-infiltrating lymphocytes (TILs), and the mutational profiles. RESULTS Seventy patients were enrolled, and 13 of 70 patients received anti-PD-1 therapy. The progression-free survival (PFS) and objective response rate (ORR) of the anti-PD-1 therapy were 4.2 months and 39%, respectively. The overall survival of patients treated with anti-PD-1 therapy (n = 13) was significantly better than those treated without anti-PD-1 therapy (n = 57) (25.2 months vs 10.9 months; P = .02). Among the 13 patients treated with anti-PD-1 therapy, 10 patients (90%) had PD-L1-negative tumors. Patients with a high density of tumoral CD8-positive TILs (≥38/mm2) had a significantly better ORR and PFS than those with a low density of tumoral CD8-positive TILs (ORR: P = .02; PFS: P = .003). Additionally, all 3 patients with TP53 mutation co-occurring with PIK3CA mutation (2 of 8 patients) or RB1 mutation (1 of 8 patients) responded to anti-PD-1 therapy. CONCLUSIONS Anti-PD-1 therapy was effective regardless of PD-L1 positivity in patients with advanced LCNEC. Our investigation might suggest that the density of tumoral CD8-positive TILs and the presence of co-occurring mutations are predictors of the efficacy of anti-PD-1 therapy in patients with advanced LCNEC.
Collapse
Affiliation(s)
- Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Daisuke Takayanagi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Katsutoshi Sekine
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Internal Medicine, Saitama City Hospital, Saitama, Japan
| | - Shintaro Kanda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Masuda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noriko Motoi
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
93
|
Lacombe C, De Rycke O, Couvelard A, Turpin A, Cazes A, Hentic O, Gounant V, Zalcman G, Ruszniewski P, Cros J, de Mestier L. Biomarkers of Response to Etoposide-Platinum Chemotherapy in Patients with Grade 3 Neuroendocrine Neoplasms. Cancers (Basel) 2021; 13:643. [PMID: 33562726 PMCID: PMC7915900 DOI: 10.3390/cancers13040643] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
Etoposide-platinum (EP) chemotherapy has long been the reference treatment for grade 3 neuroendocrine neoplasms (G3 NEN). However, G3 NEN are heterogeneous, including well-differentiated tumors (NET) and poorly differentiated large (LCNEC) or small (SCNEC) cell carcinomas, whose response to EP chemotherapy varies considerably. Our aim was to evaluate predictive biomarkers for the response to EP chemotherapy in G3 NEN. We retrospectively studied 89 patients with lung (42%) and digestive (58%) G3 NEN treated by EP chemotherapy between 2006 and 2020. All cases were centrally reviewed for cytomorphology/Ki-67 and immunohistochemistry of retinoblastoma protein (Rb)/p53/p16, analyzed using a semi-quantitative score. The absence of Rb staining (Rbinap) or the absence of very intense p53 staining (p53inap) were considered inappropriate. Rb staining was also studied as a quantitative marker, the best threshold being determined by ROC curve. Intense p16 staining (p16high) also suggested cell cycle dysregulation. Our primary endpoint was the objective response rate (ORR). We included 10 G3 NET, 31 LCNEC and 48 SCNEC, which showed ORR of 20%, 32% and 75%, respectively (NET vs. NEC, p = 0.040; LCNEC vs. SCNEC, p < 0.001). The ORR was significantly higher in NEN presenting with Rbinap (63% vs. 42%, p = 0.025) and p16high (66% vs. 35%, p = 0.006). Rb < 150 optimally identified responders (AUC = 0.657, p < 0.001). The ORR was 67% in Rb < 150 (vs. 25%, p = 0.005). On multivariate analysis, only Rb < 150 was independently associated with ORR (OR 4.16, 95% CI 1.11-15.53, p = 0.034). We confirm the heterogeneity of the response to EP treatment in G3 NEN. Rb < 150 was the best predictive biomarker for the response to EP, and p53 immunostaining had no additional value.
Collapse
Affiliation(s)
- Caroline Lacombe
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Ophélie De Rycke
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Anne Couvelard
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Anthony Turpin
- Department of Medical Oncology, Claude Huriez University Hospital, 59000 Lille, France;
| | - Aurélie Cazes
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Olivia Hentic
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
| | - Valérie Gounant
- Université de Paris, Department of Thoracic Oncology, CIC INSERM 1425, Bichat University Hospital, 75018 Paris, France; (V.G.); (G.Z.)
| | - Gérard Zalcman
- Université de Paris, Department of Thoracic Oncology, CIC INSERM 1425, Bichat University Hospital, 75018 Paris, France; (V.G.); (G.Z.)
| | - Philippe Ruszniewski
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Jérôme Cros
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Louis de Mestier
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| |
Collapse
|
94
|
Dudnik E, Kareff S, Moskovitz M, Kim C, Liu SV, Lobachov A, Gottfried T, Urban D, Zer A, Rotem O, Onn A, Wollner M, Bar J. Real-world survival outcomes with immune checkpoint inhibitors in large-cell neuroendocrine tumors of lung. J Immunother Cancer 2021; 9:e001999. [PMID: 33597218 PMCID: PMC7893659 DOI: 10.1136/jitc-2020-001999] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Little is known regarding the efficacy of immune checkpoint inhibitors (ICI) in patients with advanced large-cell neuroendocrine lung carcinoma (aLCNEC). METHODS 125 consecutive patients with aLCNEC were identified in the electronic databases of 4 participating cancer centers. The patients were divided into group A (patients who received ICI, n=41) and group B (patients who did not receive ICI, n=84). Overall survival since advanced disease diagnosis (OS DX) and OS since ICI initiation (OS ICI) were captured. RESULTS With a median follow-up of 11.8 months (mo) (IQR 7.5-17.9) and 6.0mo (IQR 3.1-10.9), 66% and 76% of patients died in groups A and B, respectively. Median OS DX was 12.4mo (95% CI 10.7 to 23.4) and 6.0mo (95% CI 4.7 to 9.4) in groups A and B, respectively (log-rank test, p=0.02). For ICI administration, HR for OS DX was 0.59 (95% CI 0.38 to 0.93, p=0.02-unadjusted), and 0.58 (95% CI 0.34 to 0.98, p=0.04-adjusted for age, Eastern Cooperative Oncology Group (ECOG) performance status (PS), presence of liver metastases and chemotherapy administration). In a propensity score matching analysis (n=74; 37 patients in each group matched for age and ECOG PS), median OS DX was 12.5 mo (95% CI 10.6 to 25.2) and 8.4 mo (95% CI 5.4 to 16.9) in matched groups A and B, respectively (log-rank test, p=0.046). OS ICI for patients receiving ICI as monotherapy (n=36) was 11.0 mo (95% CI 6.1 to 19.4). CONCLUSIONS With the limitations of retrospective design and small sample size, the results of this real-world cohort analysis suggest a positive impact of ICI on OS in aLCNEC.
Collapse
Affiliation(s)
- Elizabeth Dudnik
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Samuel Kareff
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Mor Moskovitz
- Thoracic Cancer Service, Rambam Health Care Campus, Haifa, Israel
| | - Chul Kim
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Anastasiya Lobachov
- Institute of Oncology, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| | - Teodor Gottfried
- Institute of Oncology, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| | - Damien Urban
- Institute of Oncology, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| | - Alona Zer
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Rotem
- Thoracic Cancer Service, Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Amir Onn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pulmonology Institute, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Mira Wollner
- Thoracic Cancer Service, Rambam Health Care Campus, Haifa, Israel
| | - Jair Bar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Oncology, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| |
Collapse
|
95
|
Mograbi B, Heeke S, Hofman P. The Importance of STK11/ LKB1 Assessment in Non-Small Cell Lung Carcinomas. Diagnostics (Basel) 2021; 11:196. [PMID: 33572782 PMCID: PMC7912095 DOI: 10.3390/diagnostics11020196] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the recent implementation of immunotherapy as a single treatment or in combination with chemotherapy for first-line treatment of advanced non-small cell lung cancer (NSCLC), many patients do not benefit from this regimen due to primary treatment resistance or toxicity. Consequently, there is an urgent need to develop efficient biomarkers that can select patients who will benefit from immunotherapy thereby providing the appropriate treatment and avoiding toxicity. One of the biomarkers recently described for the stratification of NSCLC patients undergoing immunotherapy are mutations in STK11/LKB1, which are often associated with a lack of response to immunotherapy in some patients. Therefore, the purpose of this review is to describe the different cellular mechanisms associated with STK11/LKB1 mutations, which may explain the lack of response to immunotherapy. Moreover the review addresses the co-occurrence of additional mutations that may influence the response to immunotherapy and the current clinical studies that have further explored STK11/LKB1 as a predictive biomarker. Additionally this work includes the opportunities and limitations to look for the STK11/LKB1 status in the therapeutic strategy for NSCLC patients.
Collapse
Affiliation(s)
- Baharia Mograbi
- Centre Antoine Lacassagne, CNRS, FHU OncoAge, Team 4, INSERM, IRCAN, Université Côte d’Azur, 06000 Nice, France;
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Paul Hofman
- Centre Antoine Lacassagne, CNRS, FHU OncoAge, Team 4, INSERM, IRCAN, Université Côte d’Azur, 06000 Nice, France;
- CHU Nice, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Pasteur Hospital, Université Côte d’Azur, 06000 Nice, France
- CHU Nice, FHU OncoAge, Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, 06000 Nice, France
| |
Collapse
|
96
|
Milione M, Maisonneuve P, Grillo F, Mangogna A, Centonze G, Prinzi N, Pusceddu S, Garzone G, Cattaneo L, Busico A, Bossi P, Spaggiari P, Pellegrinelli A, Del Gobbo A, Ferrero S, Kankava K, Pruneri G, Rolli L, Roca E, Bercich L, Tironi A, Benvenuti MR, Gallazzi MS, Romano R, Berruti A, Pastorino U, Capella C. Ki-67 Index of 55% Distinguishes Two Groups of Bronchopulmonary Pure and Composite Large Cell Neuroendocrine Carcinomas with Distinct Prognosis. Neuroendocrinology 2021; 111:475-489. [PMID: 32365350 DOI: 10.1159/000508376] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/01/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Little information is available concerning prognostic factors for bronchopulmonary large cell neuroendocrine carcinomas (BP-LCNECs) and even less is known about combined LCNECs (Co-LCNECs). We investigated whether an integrated morphological, immunohistochemical, and molecular approach could be used for their prognostic evaluation. METHODS Morphological (including combined features), proliferative (mitotic count/Ki-67 index), immunohistochemical (napsin A, p40, TTF-1, CD44, OTP, SSTR2A, SSTR5, mASH1, p53, RB1, and MDM2), and genomic (TP53, RB1, ATM, JAK2, KRAS, and STK11) findings were analyzed in BP-LCNECs from 5 Italian centers, and correlated with overall survival (OS). The Ki-67 index was expressed as the percentage of positive cells in hot spots as indicated in the WHO 2019 Digestive System Tumors and, for Co-LCNECs, the Ki-67 index was evaluated only in the LCNEC component. RESULTS A total of 111 LCNECs were distinguished into 70 pure LCNECs, 35 Co-LCNECs (27 with adenocarcinoma [ADC] and 8 with squamous cell carcinoma [SqCC]), and 6 LCNECs with only napsin A immunoreactivity. The Ki-67 index cutoff at 55% evaluated in the neuroendocrine component was the most powerful predictor of OS (log-rank p = 0.0001) in all LCNECs; 34 cases had a Ki-67 index <55% (LCNEC-A) and 77 had a Ki-67 index ≥55% (LCNEC-B). Statistically significant differences in OS (log-rank p = 0.0001) were also observed between pure and Co-LCNECs. A significant difference in OS was found between pure LCNECs-A and Co-LCNECs-A (p < 0.05) but not between pure LCNECs-B and Co-LCNECs-B. Co-LCNEC-ADC and LCNEC napsin A+ cases had longer OS than pure LCNEC and Co-LCNEC-SqCC cases (log-rank p = 0.0001). On multivariable analysis, tumor location, pure versus combined features, and napsin A, but no single gene mutation, were significantly associated with OS after adjustment for Ki-67 index and study center (p < 0.05). CONCLUSIONS The Ki-67 proliferation index and the morphological characterization of combined features in LCNECs seem to be important tools for predicting clinical outcome in BP-LCNECs.
Collapse
Affiliation(s)
- Massimo Milione
- First Division of Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy,
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Grillo
- Unit of Pathology, Department of Surgical Sciences and Integrated Diagnostics, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessandro Mangogna
- Unit of Pathology, Clinical Department of Medical, Surgical and Health Science, University of Trieste, Ospedale di Cattinara, Trieste, Italy
| | - Giovanni Centonze
- First Division of Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Natalie Prinzi
- Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Pusceddu
- Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanna Garzone
- First Division of Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Cattaneo
- First Division of Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Adele Busico
- 2nd Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Paola Bossi
- Pathology Department, Humanitas Clinical and Research Center, Humanitas Milan ENETS Center of Excellence, Milan, Italy
| | - Paola Spaggiari
- Pathology Department, Humanitas Clinical and Research Center, Humanitas Milan ENETS Center of Excellence, Milan, Italy
| | - Alessio Pellegrinelli
- Department of Pathology, ASST Franciacorta, Mellino Mellini Hospital, Brescia, Italy
| | - Alessandro Del Gobbo
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Ketevani Kankava
- Teaching, Scientific and Diagnostic Pathology Laboratory, Tbilisi State Medical University, Tbilisi, Georgia
| | - Giancarlo Pruneri
- 2nd Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
- School of Medicine, University of Milan, Milan, Italy
| | - Luigi Rolli
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Elisa Roca
- Medical Oncology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Luisa Bercich
- Department of Pathology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Andrea Tironi
- Department of Pathology, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mauro Roberto Benvenuti
- Thoracic Surgery Unit, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Maria Sole Gallazzi
- Thoracic Surgery Unit, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Rosalia Romano
- Thoracic Surgery Unit, Department of Medical and Surgical Specialties Radiological Sciences and Public Health, Medical Oncology, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Science and Public Health, University of Brescia, Brescia, Italy
| | - Ugo Pastorino
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Capella
- Unit of Pathology, Department of Medicine and Surgery and Research Center for the Study of Hereditary and Familial tumors, University of Insubria, Varese, Italy
| |
Collapse
|
97
|
Cai C, Zeng Q, Zhou G, Mu X. Identification of novel transcription factor-microRNA-mRNA co-regulatory networks in pulmonary large-cell neuroendocrine carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:133. [PMID: 33569435 PMCID: PMC7867924 DOI: 10.21037/atm-20-7759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Large cell neuroendocrine carcinoma (LCNEC) of the lung is a rare neuroendocrine neoplasm. Previous studies have shown that microRNAs (miRNAs) are widely involved in tumor regulation through targeting critical genes. However, it is unclear which miRNAs play vital roles in the pathogenesis of LCNEC, and how they interact with transcription factors (TFs) to regulate cancer-related genes. Methods To determine the novel TF-miRNA-target gene feed-forward loop (FFL) model of LCNEC, we integrated multi-omics data from Gene Expression Omnibus (GEO), Transcriptional Regulatory Relationships Unraveled by Sentence-Based Text Mining (TRRUST), Transcriptional Regulatory Element Database (TRED), and The experimentally validated microRNA-target interactions database (miRTarBase database). First, expression profile datasets for mRNAs (GSE1037) and miRNAs (GSE19945) were downloaded from the GEO database. Overlapping differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) were identified through integrative analysis. The target genes of the FFL were obtained from the miRTarBase database, and the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses were performed on the target genes. Then, we screened for key miRNAs in the FFL and performed gene regulatory network analysis based on key miRNAs. Finally, the TF-miRNA-target gene FFLs were constructed by the hypergeometric test. Results A total of 343 DEGs and 60 DEMs were identified in LCNEC tissues compared to normal tissues, including 210 down-regulated and 133 up-regulated genes, and 29 down-regulated and 31 up-regulated miRNAs. Finally, the regulatory network of TF-miRNA-target gene was established. The key regulatory network modules included ETS1-miR195-CD36, TAOK1-miR7-1-3P-GRIA1, E2F3-miR195-CD36, and TEAD1-miR30A-CTHRC1. Conclusions We constructed the TF-miRNA-target gene regulatory network, which is helpful for understanding the complex LCNEC regulatory mechanisms.
Collapse
Affiliation(s)
- Cunliang Cai
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Qianli Zeng
- The South China Center for Innovative Pharmaceuticals, Guangzhou, China
| | - Guiliang Zhou
- The South China Center for Innovative Pharmaceuticals, Guangzhou, China
| | - Xiangdong Mu
- Department of Respiratory and Critical Care Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
98
|
Komisarof J, Qiu H, Velez MJ, Mulford D. Anterior mediastinal large cell neuroendocrine carcinoma with elevated AFP: A case report and review. Mol Clin Oncol 2020; 14:34. [PMID: 33414915 PMCID: PMC7783711 DOI: 10.3892/mco.2020.2196] [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: 02/06/2020] [Accepted: 11/18/2020] [Indexed: 11/05/2022] Open
Abstract
Large cell neuroendocrine carcinoma (LCNEC) is a rare and aggressive cancer that typically presents in the lung. The current case report describes a 56 year old male who presented to Strong Memorial Hospital with progressive dyspnea and was revealed to have a large anterior mediastinal tumor with metastases to axillary, hilar and mediastinal lymph nodes. Tumor marker results revealed an elevated plasma level of α-fetoprotein (AFP), which initially pointed towards a diagnosis of teratoma, but the tumor stained positive for neuroendocrine markers CD56, chromogranin, and synaptophysin on biopsy, consistent with LCNEC. AFP-positive tumor cells were identified, and no alternate cause for the elevated AFP was identified. The patient underwent genetic testing revealing the tumor to be ALK, ROS1, KRAS, BRAF and EGFR wild type. The patient received 6 cycles of chemotherapy with cisplatin (80 mg/m2) and etoposide (100 mg/m2) and then radiation with an initial minor response. The patients course was complicated by the development of superior vena cava syndrome requiring emergency stenting. The results of the current case suggest that AFP may be worthy of further exploration as a potential tumor marker in LCNEC.
Collapse
Affiliation(s)
- Justin Komisarof
- Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Haoming Qiu
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Moises J Velez
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah Mulford
- Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
99
|
Molecular Characterization of Neuroendocrine Carcinomas of the Endometrium: Representation in All 4 TCGA Groups. Am J Surg Pathol 2020; 44:1541-1548. [PMID: 32773531 DOI: 10.1097/pas.0000000000001560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
High-grade neuroendocrine carcinomas (NEC) of the endometrium are rare and account for <1% of all endometrial carcinomas. Both small cell neuroendocrine carcinoma (SCNEC) and large cell neuroendocrine carcinoma (LCNEC) morphologies have been reported. Little is known regarding the molecular features of endometrial NEC including how they compare to pulmonary NEC (the most common site for these neoplasms) and the more common endometrial carcinoma histotypes. In this study, we investigated the molecular alterations in a series of endometrial NEC using a targeted next generation sequencing panel (Oncopanel). Fourteen NEC were sequenced; pure NEC (n=4) and mixed (n=10) with endometrioid adenocarcinoma (n=9) or carcinosarcoma (n=1). The NEC components of mixed tumors comprised LCNEC (n=6) and SCNEC (n=4). The 4 pure NEC comprised LCNEC (n=2) and SCNEC (n=2). Molecular analysis classified tumors into the 4 The Cancer Genome Atlas groups: (1) POLE-mutated/ultramutated (1/14; 7%), (2) microsatellite instability/hypermutated (6/14; 43%), (3) TP53 mutated/copy number high (2/14; 14%), or (4) no specific molecular profile (5/14; 36%). Overall, 50% of cases were ultramutated or hypermutated. In 8 cases of mixed carcinomas, the different histologic components were macrodissected and separately sequenced; molecular alterations were nearly identical among the 2 components, with the non-NEC component harboring slightly increased tumor mutational burden. Only 2 carcinomas (both with pure SCNEC morphology) had a molecular profile that would be expected in typical pulmonary SCNEC (RB1 deletion and TP53 mutations). Our findings, similar to data from NECs of other anatomic sites, suggest that the molecular context may be important when selecting therapies for women with endometrial NEC. Immune checkpoint inhibition may be a reasonable approach to treatment of microsatellite instability-NEC and we thus recommend that all endometrial NEC be tested for mismatch repair abnormalities, either molecularly or by mismatch repair protein immunohistochemistry.
Collapse
|
100
|
Hermans BCM, Derks JL, Moonen L, Habraken CHJ, der Thüsen JV, Hillen LM, Speel EJM, Dingemans AMC. Pulmonary neuroendocrine neoplasms with well differentiated morphology and high proliferative activity: illustrated by a case series and review of the literature. Lung Cancer 2020; 150:152-158. [PMID: 33171403 DOI: 10.1016/j.lungcan.2020.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 09/07/2020] [Accepted: 10/16/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Pulmonary neuroendocrine neoplasms (NENs) are subdivided in carcinoids and neuroendocrine carcinomas (small cell lung carcinoma and large cell neuroendocrine carcinoma (LCNEC)), based on the presence of necrosis and mitotic index (MI). However, it is unclear if tumors with well differentiated morphology but high proliferation rate should be regarded as LCNEC or as high grade carcinoids. In previous case series, a longer overall survival then expected in LCNEC has been suggested. We describe 7 of those cases analyzed for pRb expression and overall survival. MATERIAL AND METHODS Cases with well differentiated morphology, but MI > 10/2mm2 and/or Ki-67 proliferation index >20% were selected based on pathology reports of consecutive NENs in our university medical center (Maastricht UMC+, 2007-2018) and confirmed by pathological review. Immunohistochemistry was performed to assess pRb expression. RESULTS Seven stage IV cases were included in this study. Median overall survival was 8 months (95% confidence interval 5-11 months). Cases with well differentiated morphology and preserved pRb expression (4/7) had a median overall survival of 45 months. CONCLUSION A subgroup of pulmonary NENs with well differentiated morphology but high proliferation rate likely exists. pRb staining might be helpful to predict prognosis, but clinical relevance remains to be studied.
Collapse
Affiliation(s)
- B C M Hermans
- Department of Pulmonary Diseases, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - J L Derks
- Department of Pulmonary Diseases, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - L Moonen
- Department of Pathology, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - C H J Habraken
- Department of Pulmonary Diseases, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - J von der Thüsen
- Department of Pathology, Erasmus Medical Centre, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| | - L M Hillen
- Department of Pathology, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - E J M Speel
- Department of Pathology, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - A-M C Dingemans
- Department of Pulmonary Diseases, GROW school for Oncology & Developmental Biology, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands; Department of Pulmonary Diseases, Erasmus Medical Centre, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|