1
|
Luo Z, Yan X, Liu Y, Nan F, Lei Y, Ren Y, Li L. Prognostic significance of Ki-67 in assessing the risk of progression, relapse or metastasis in pheochromocytomas and paragangliomas. Ann Med 2025; 57:2478312. [PMID: 40079941 PMCID: PMC11984564 DOI: 10.1080/07853890.2025.2478312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 03/15/2025] Open
Abstract
INTRODUCTION Since the Fourth edition of the WHO classification, PPGLs have been recognized for their metastatic potential, though no clear features can accurately predict this behavior. The prognostic value of Ki-67 in assessing the risk of progression, relapse, or metastasis in PPGLs remains debated. METHODS This cohort study included 501 patients diagnosed with PPGLs at the First Hospital of Jilin University between 2000 and 2022, with clinical data, treatment details, pathological indicators, and germline gene test results collected. Bulk sequencing was performed on formalin-fixed paraffin-embedded (FFPE) primary tumor samples from 87 patients. Progression-free survival (PFS) was analyzed using multivariable Cox regression. RESULTS Among the 119 enrolled patients with PPGLs, the average age was 45.7 ± 14.0 years, and the median follow-up time was 46 months. A significant finding was the high expression of CDK1, a gene known to be significantly associated with the metastatic risk of PPGLs, in samples with Ki-67 ≥ 3% (p < 0.0001). More importantly, patients with PPGLs and a Ki-67 level ≥ 3% had a 3.59-fold higher risk of progression, relapse or metastasis compared to those with Ki-67 < 3% (HR = 4.59, 95% CI: 1.06-11.95), after adjusting for all confounding factors. In the composite model, the addition of Ki-67 enhanced the predictive ability of the combined model of SDHB, primary site, tumor size, and invade neighboring tissue (AUC = 0.888, 95% CI: 0.808-0.967 vs. AUC = 0.874, 95% CI: 0.783-0.965). CONCLUSION A Ki-67 level ≥ 3% is associated with an increased risk of progression, relapse or metastasis in patients with PPGLs.
Collapse
Affiliation(s)
- Zilan Luo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Yan
- Pathology Department, The First Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Fengrui Nan
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Yuhong Lei
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Yuan Ren
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Zhan Y, Zheng F, Liao S, Lu X, Cao F, Huang T, Yang X, Han Y, Zeng L, Li L, Huang W. Epstein-Barr virus-positive small cell neuroendocrine carcinoma of the nasopharynx: A clinicopathologic study of 7 cases and literature review. Hum Pathol 2025; 158:105787. [PMID: 40354885 DOI: 10.1016/j.humpath.2025.105787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/03/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Epstein-Barr virus (EBV)-positive small cell neuroendocrine carcinoma (NEC) of the nasopharynx is a rare entity with poorly characterized clinicopathological and prognostic features. In this study, seven cases of EBV-positive nasopharyngeal small cell NEC will be compared to 6 EBV-negative cases of nasopharyngeal small cell NEC. The overall EBV-positive rate, defined by EBV-encoded small RNAs (EBERs) in situ hybridization of nasopharyngeal small cell NEC, was 53.8 % (7/13). The patients' age ranged from 38 to 73 years, with a median age of 56 years. There was a large preponderance of males, with a male-to-female ratio of 6:1. In the present study, EBV-positive nasopharyngeal small cell NEC had a significantly higher expression of POU2F3 (85.7 %, 6/7) than EBV-negative nasopharyngeal small cell NEC (16.7 %, 1/6). RNA sequencing revealed that EBV-positive nasopharyngeal small cell NEC was distinct from EBV-negative nasopharyngeal small cell NEC in the control group. There was no statistically significant difference in overall survival between patients with EBV-positive and EBV-negative nasopharyngeal small cell NEC in the present cohort. These findings suggest that EBV-positive nasopharyngeal small cell NEC may be closely associated with POU2F3. In addition, POU2F3 and ASCL1 may play important roles in the tumorigenesis of EBV-positive nasopharyngeal small cell NEC.
Collapse
Affiliation(s)
- Yuan Zhan
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fei Zheng
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Shousheng Liao
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Xiangtong Lu
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Fang Cao
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Tao Huang
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Xuan Yang
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Yue Han
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Lei Zeng
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Lixiang Li
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Wenyong Huang
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, China.
| |
Collapse
|
3
|
Fukushima T, Togasaki K, Hamamoto J, Emoto K, Ebisudani T, Mitsuishi A, Sugihara K, Shinozaki T, Okada M, Saito A, Takaoka H, Ito F, Shigematsu L, Ohta Y, Takahashi S, Matano M, Kurebayashi Y, Ohgino K, Sato T, Kawada I, Asakura K, Hishida T, Asamura H, Ikemura S, Terai H, Soejima K, Oda M, Fujii M, Fukunaga K, Yasuda H, Sato T. An organoid library unveils subtype-specific IGF-1 dependency via a YAP-AP1 axis in human small cell lung cancer. NATURE CANCER 2025:10.1038/s43018-025-00945-y. [PMID: 40307487 DOI: 10.1038/s43018-025-00945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/07/2025] [Indexed: 05/02/2025]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with limited therapeutic advancements. Although SCLC has recently been classified into four molecular subtypes, subtype-specific therapies are still lacking. Here, we established 40 patient-derived SCLC organoid lines with predominant TP53 and RB1 alterations and rare targetable genetic lesions. Transcriptome profiling divided the SCLC organoids into neuroendocrine (NE)-type SCLC and non-NE-type SCLC, with the latter characterized by YAP1 or POU2F3 expression. NE-type SCLC organoids grew independent of alveolar niche factors, whereas non-NE-type SCLC organoids relied on insulin-like growth factor (IGF)-1-driven YAP1 and AP1 activation. Therapeutic targeting of IGF-1, YAP1 and AP1 effectively suppressed the growth of non-NE-type organoids. Co-knockout of TP53 and RB1 in human alveolar cells altered their lineage toward the airway epithelium-like fate and conferred IGF-1 dependency, validating the subtype-phenotype connection. Our SCLC organoid library represents a valuable resource for developing biology-based therapies and has the potential to reshape the drug discovery landscape.
Collapse
Affiliation(s)
- Takahiro Fukushima
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Division of Gastroenterology Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Katsura Emoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Ebisudani
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akifumi Mitsuishi
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kai Sugihara
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Taro Shinozaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Masahiko Okada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Ayaka Saito
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Hatsuyo Takaoka
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Fumimaro Ito
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Lisa Shigematsu
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ohgino
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Takashi Sato
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ichiro Kawada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Keisuke Asakura
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyuki Hishida
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hisao Asamura
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shinnosuke Ikemura
- Department of Pulmonary Medicine, Faculty of Medicine University of Yamanashi, Yamanashi, Japan
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kenzo Soejima
- Department of Pulmonary Medicine, Faculty of Medicine University of Yamanashi, Yamanashi, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan.
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
4
|
Cognigni V, Toscani I, D’Agnelli S, Pecci F, Righi L, Berardi R, Tiseo M. Molecular heterogeneity of small cell lung cancer and new therapeutic possibilities: a narrative review of the literature. Transl Lung Cancer Res 2025; 14:1441-1455. [PMID: 40386726 PMCID: PMC12082233 DOI: 10.21037/tlcr-24-755] [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: 09/14/2024] [Accepted: 01/08/2025] [Indexed: 05/20/2025]
Abstract
Background and Objective Small cell lung cancer (SCLC) is an aggressive disease commonly occurring in individuals with a history of heavy smoking. Despite recent approvals of chemotherapy and immunotherapy in the first-line treatment of extensive-stage SCLC, it maintains a poor prognosis. Moreover, only a small percentage of patients benefits from the addition of immunotherapy to platinum-based chemotherapy. The lack of significant progress in therapeutic options unrevealed the urgent need for a deeper understanding of tumor biology and easy-to-use predictive biomarkers, aiming to better tailor the treatment strategy. The aim of this review is to summarize recent evidence about the biology, molecular heterogeneity, as well as tumor microenvironment (TME) of SCLC and their forefront therapeutic implications. Methods A literature search was conducted using PubMed, focusing on articles published in English from 1981 to October 2024. Studies on SCLC biology and subclassification were selected for further analysis and integrated in the current narrative review. Key Content and Findings SCLC entity implies four distinct molecular subtypes based on transcription factors expression, specifically achaete-scute homolog 1 (ASCL1), neurogenic differentiation 1 (NEUROD1), POU class 2 homeobox 3 (POU2F3), and yes-associated protein 1 (YAP1), reflecting the tumor heterogeneity in terms of gene expression, transcriptional profiles, immune infiltration, and treatment sensitivity. Recently, a new subgroup, "SCLC-I", has been proposed to replace the YAP1 subtype, showing higher responsiveness to immunotherapy. The TME, implying immune cell infiltration and their interactions with cancer cells, plays a crucial role in determining SCLC's sensitivity to immunotherapy. Conclusions Advances in SCLC molecular characterization and the development of targeted therapies against specific molecular pathways might improve patients' clinical outcome, supporting a more personalized approach to this complex disease.
Collapse
Affiliation(s)
- Valeria Cognigni
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Ilaria Toscani
- Medical Oncology Unit, Hospital of Piacenza, Piacenza, Italy
| | - Simona D’Agnelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Federica Pecci
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Luisella Righi
- Pathology Unit, Department of Oncology, University of Torino at San Luigi Hospital, Orbassano, Italy
| | - Rossana Berardi
- Department of Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
5
|
Catozzi A, Peiris Pagès M, Humphrey S, Revill M, Morgan D, Roebuck J, Chen Y, Davies-Williams B, Brennan K, Mukarram Hossain ASM, Makeev VJ, Satia K, Sfyri PP, Galvin M, Coles D, Lallo A, Pearce SP, Kerr A, Priest L, Foy V, Carter M, Caeser R, Chan JM, Rudin CM, Blackhall F, Frese KK, Dive C, Simpson KL. Functional characterization of the ATOH1 molecular subtype indicates a pro-metastatic role in small cell lung cancer. Cell Rep 2025; 44:115603. [PMID: 40305287 DOI: 10.1016/j.celrep.2025.115603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/09/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Molecular subtypes of small cell lung cancer (SCLC) have been described based on differential expression of the transcription factors (TFs) ASCL1, NEUROD1, and POU2F3 and immune-related genes. We previously reported an additional subtype based on expression of the neurogenic TF ATOH1 within our SCLC circulating tumor cell-derived explant (CDX) model biobank. Here, we show that ATOH1 protein is detected in 7 of 81 preclinical models and 16 of 102 clinical samples of SCLC. In CDX models, ATOH1 directly regulates neurogenesis and differentiation programs, consistent with roles in normal tissues. In ex vivo cultures of ATOH1+ CDXs, ATOH1 is required for cell survival. In vivo, ATOH1 depletion slows tumor growth and suppresses liver metastasis. Our data validate ATOH1 as a bona fide lineage-defining TF of SCLC with cell survival and pro-metastatic functions. Further investigation exploring ATOH1-driven vulnerabilities for targeted treatment with predictive biomarkers is warranted.
Collapse
Affiliation(s)
- Alessia Catozzi
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Maria Peiris Pagès
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Sam Humphrey
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Mitchell Revill
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Derrick Morgan
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Jordan Roebuck
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Yitao Chen
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Bethan Davies-Williams
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Kevin Brennan
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - A S Md Mukarram Hossain
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Vsevolod J Makeev
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Karishma Satia
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Pagona P Sfyri
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Melanie Galvin
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Darryl Coles
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Alice Lallo
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Simon P Pearce
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Alastair Kerr
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Lynsey Priest
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Victoria Foy
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Mathew Carter
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Rebecca Caeser
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joseph M Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fiona Blackhall
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M20 4BX, UK
| | - Kristopher K Frese
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK.
| | - Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
6
|
Kitahama K, Shigematsu Y, Sugawara E, Amori M, Amori G, Saito R, Ohmoto A, Yonese J, Takeuchi K, Inamura K. Clinicopathological characteristics of transcription factor-defined subtypes in bladder small cell carcinoma. BMC Cancer 2025; 25:766. [PMID: 40275187 PMCID: PMC12020276 DOI: 10.1186/s12885-025-14157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Small cell carcinoma (SmCC) of the bladder is a rare and aggressive malignancy. Characterizing transcription factor (TF)-defined subtypes may provide insights into its biology and inform targeted therapies. This study investigates lineage-specific TF expression in bladder SmCC, its association with clinicopathological features, and comparisons with prostate SmCC. METHODS A retrospective analysis was conducted on 9 cases of bladder SmCC and 6 cases of prostate SmCC diagnosed at a single cancer hospital in Japan. Immunohistochemistry was performed for lineage-specific TFs (ASCL1, NEUROD1, POU2F3, and YAP1) and neuroendocrine and other markers. Statistical comparisons were made using Fisher's exact test and independent samples t-tests. RESULTS Combined SmCC morphology, including urothelial carcinoma (UC) (5 cases) and adenocarcinoma (2 cases), was more frequent in bladder SmCC than in prostate SmCC (78% [7 of 9 cases] vs. 17% [1 of 6 cases], p = 0.041). NEUROD1 was more frequently expressed in bladder SmCC than in prostate SmCC (67% [6 of 9 cases] vs. 0% [0 of 6 cases]; p = 0.028). NEUROD1 expression was more frequent in combined SmCC and UC bladder tumors than in other bladder SmCC tumors (100% [5 of 5 cases] vs. 25% [1 of 4 cases], p = 0.048). Conversely, HNF4A expression was absent in all combined SmCC and UC bladder tumors (0 of 5) but present in 75% (3 of 4) of other bladder SmCC tumors (p = 0.048). In 2 cases of bladder SmCC, NEUROD1 and POU2F3 were expressed in a mutually exclusive manner, with neuroendocrine markers expressed only in the NEUROD1-expressing component. CONCLUSIONS NEUROD1 is characteristically expressed in bladder SmCC, especially in SmCC combined with UC, suggesting a distinct phenotype from prostate SmCC. These findings highlight the potential for TF-based classification to improve diagnostic accuracy and inform therapeutic strategies.
Collapse
Affiliation(s)
- Keiichiro Kitahama
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Pathology, Kyorin University School of Medicine, Tokyo, Japan
| | - Yasuyuki Shigematsu
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Emiko Sugawara
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mahmut Amori
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Gulanbar Amori
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan
| | - Rumiko Saito
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Clinical Chemotherapy, The Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Graduate School of Engineering, Chiba Institute of Technology, Chiba, Japan
| | - Akihiro Ohmoto
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Division of Tumor Pathology, Jichi Medical University, Shimotsuke, Japan.
| |
Collapse
|
7
|
Zhang Z, Wu X, Bao S, Sun X, Yang F, Zhang Y, Yang Z, Zhang L, Chen R, Xing P, Li J, Zhou M, Yang L. Proteogenomic Characterization of High-Grade Lung Neuroendocrine Carcinoma Deciphers Molecular Diversity and Potential Biomarkers of Different Histological Subtypes in Chinese Population. RESEARCH (WASHINGTON, D.C.) 2025; 8:0671. [PMID: 40230612 PMCID: PMC11994885 DOI: 10.34133/research.0671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/18/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025]
Abstract
High-grade lung neuroendocrine carcinomas (Lu-NECs) are clinically refractory malignancies with poor prognosis and limited therapeutic advances. The biological and molecular features underlying the histological heterogeneity of Lu-NECs are not fully understood. In this study, we present a multi-omics integration of whole-exome sequencing and deep proteomic profiling in 93 Chinese Lu-NECs to establish the first comprehensive proteogenomic atlas of this disease spectrum. Our analyses revealed a high degree of mutational concordance among the subtypes at the genomic level; however, distinct proteomic profiles enabled a clear differentiation of histological subtypes, unveiling subtype-specific molecular and biological features related to tumor metabolism, immunity, and proliferation. Furthermore, RB1 mutations confer divergent prognostic effects through subtype-specific cis- and trans-proteomic regulation. In addition, we identified potential protein biomarkers for histological subtype classification and risk stratification, which were validated by immunohistochemistry in an independent cohort. This study provides a valuable proteogenomic resource and insight into Lu-NEC heterogeneity.
Collapse
Affiliation(s)
- Zicheng Zhang
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Xi Wu
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Siqi Bao
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Fan Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Yibo Zhang
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Zijian Yang
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Liujin Zhang
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Ruanqi Chen
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Puyuan Xing
- Department of Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Junling Li
- Department of Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| | - Meng Zhou
- School of Biomedical Engineering,
Wenzhou Medical University, Wenzhou 325027, P. R. China
| | - Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P. R. China
| |
Collapse
|
8
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025:10.1038/s41568-025-00803-0. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
9
|
Morin A, Chu CP, Pavlidis P. Identifying reproducible transcription regulator coexpression patterns with single cell transcriptomics. PLoS Comput Biol 2025; 21:e1012962. [PMID: 40257984 PMCID: PMC12011263 DOI: 10.1371/journal.pcbi.1012962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/13/2025] [Indexed: 04/23/2025] Open
Abstract
The proliferation of single cell transcriptomics has potentiated our ability to unveil patterns that reflect dynamic cellular processes such as the regulation of gene transcription. In this study, we leverage a broad collection of single cell RNA-seq data to identify the gene partners whose expression is most coordinated with each human and mouse transcription regulator (TR). We assembled 120 human and 103 mouse scRNA-seq datasets from the literature (>28 million cells), constructing a single cell coexpression network for each. We aimed to understand the consistency of TR coexpression profiles across a broad sampling of biological contexts, rather than examine the preservation of context-specific signals. Our workflow therefore explicitly prioritizes the patterns that are most reproducible across cell types. Towards this goal, we characterize the similarity of each TR's coexpression within and across species. We create single cell coexpression rankings for each TR, demonstrating that this aggregated information recovers literature curated targets on par with ChIP-seq data. We then combine the coexpression and ChIP-seq information to identify candidate regulatory interactions supported across methods and species. Finally, we highlight interactions for the important neural TR ASCL1 to demonstrate how our compiled information can be adopted for community use.
Collapse
Affiliation(s)
- Alexander Morin
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ching Pan Chu
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Pavlíčková K, Hojný J, Waldauf P, Švajdler M, Dundr P, Fabian P, Krkavcová E, Dvořák J, Michálková R, Zambo IS, Hájková N, Flídrová M, Laco J, Hornychová H, Delongová P, Škarda J, Hrudka J, Matěj R. Molecular and Immunohistochemical Classification of Extrapulmonary Small Cell Neuroendocrine Carcinomas: A Study of 181 Cases. J Transl Med 2025; 105:104093. [PMID: 39826683 DOI: 10.1016/j.labinv.2025.104093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/28/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Extrapulmonary small cell neuroendocrine carcinoma (EP-SCNC) is a rare malignancy with a poor prognosis. Most patients with EP-SCNC have metastatic disease upon presentation, and their average overall survival (OS) is less than 12 months. Our study aimed to conduct a complex analysis of EP-SCNC. One hundred eighty-one EP-SCNC tissue samples were subjected to a complex analysis. One hundred fifty-five tumors were pure EP-SCNC, whereas 26 were combined tumors. Immunohistochemistry for ASCL1, NEUROD1, YAP1, POU2F3, Rb1, p53, cyclin D1, p16, PTEN, DLL3, PD-L1, CD56, synaptophysin, chromogranin A, and INSM1 was performed, and 128 samples were analyzed molecularly using next-generation sequencing, comprising DNA and RNA analyses. Detailed results on immunohistochemical and molecular analyses were provided for each primary origin of EP-SCNC separately. Median survival for the whole cohort of patients was 8.94 months. Patient age (≥70 years), tumor mutational burden <15, and TP53 and BRCA2 mutations were negative prognostic factors. High expression of ASCL-1 was associated with shorter OS, whereas high expression of YAP1 was associated with longer OS. Patients with genitourinary tumors had significantly better OS than those with gastrointestinal tract EP-SCNC tumors. Rb1 expression loss was detected more often in genitourinary tract SCNCs. In contrast, p16 overexpression was found more often in genitourinary tract SCNCs. POU2F3 expression was detected more often in combined tumors, whereas NEUROD1 was detected more often in pure EP-SCNC. Regarding "druggable markers," DLL3 was expressed in 66% of tumors and PD-L1 in 17.4%. Detailed analyses of different prognostic and predictive markers are needed to better understand EP-SCNC biology and create more personalized therapy to improve patient prognosis.
Collapse
MESH Headings
- Humans
- Male
- Female
- Aged
- Middle Aged
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/classification
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/metabolism
- Immunohistochemistry
- Adult
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/analysis
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/classification
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/mortality
- Prognosis
Collapse
Affiliation(s)
- Klára Pavlíčková
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Hojný
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petr Waldauf
- Department of Anesthesia and Intensive Care, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Marián Švajdler
- Bioptická laboratoř s.r.o. and Šikl's Department of Pathology, Charles University, Medical Faculty in Pilsen, Pilsen, Czech Republic
| | - Pavel Dundr
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Fabian
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Eva Krkavcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jiří Dvořák
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Romana Michálková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Iva Staniczková Zambo
- 1st Institute of Pathologic Anatomy, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Nikola Hájková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Miroslava Flídrová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Kralove, Hradec Králové, Czech Republic
| | - Helena Hornychová
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Kralove, Hradec Králové, Czech Republic
| | - Patricie Delongová
- Department of Clinical and Molecular Pathology and Medical Genetics, University Hospital Ostrava and Faculty of Medicine University of Ostrava, Ostrava, Czech Republic
| | - Jozef Škarda
- Department of Clinical and Molecular Pathology and Medical Genetics, University Hospital Ostrava and Faculty of Medicine University of Ostrava, Ostrava, Czech Republic
| | - Jan Hrudka
- Department of Pathology, University Hospital Kralovske Vinohrady, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radoslav Matěj
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Department of Pathology, University Hospital Kralovske Vinohrady, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
11
|
Xu J, Zhang W, Xie F, Wang C, Cheng F, Rao R, Chen Y, Zhang L, Wen W, Zhao Z, Yuan J, Zheng Y, Yu Z. Prognostic significance of ALK high expression in SCLC: a 9-year cohort analysis. Front Oncol 2025; 15:1530339. [PMID: 40201354 PMCID: PMC11975910 DOI: 10.3389/fonc.2025.1530339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose The aim of this study was to investigate the prognostic value of the abnormal expression of anaplastic lymphoma kinase (ALK) protein in patients with small cell lung cancer (SCLC) based on 9-year data from our center. Methods A retrospective cohort study was conducted to assess the clinical outcomes of patients with ALK-positive SCLC diagnosed in our hospital over the past 9 years. We used public databases to analyze the expression of ALK in pan-cancer and its prognostic value and analyzed the correlation between ALK and SCLC prognosis-related genes. Results A total of 685 patients diagnosed with SCLC underwent ALK testing, and 59 patients were identified to have abnormal expression of the ALK protein, with 10 cases showing strong expression, 14 cases displaying moderate expression, and 35 cases exhibiting weak expression. The median age of the ALK-positive cohort was 64 years (range: 58-70 years), 91.5% (54/59) were male, 61.0% (36/59) were smokers, and the median overall survival (mOS) was 7.0 months (95% CI: 4.5-9.5 months). Within this cohort, the mOS for the ALK (+) subgroup was 4.0 months (95% CI: 2.9-5.1 months), the mOS for the ALK (++) subgroup was 10.0 months (95% CI: 4.9-15.1 months), and the mOS for the ALK (+++) subgroup was 12.0 months (95% CI: 7.4-16.6 months). Kaplan-Meier revealed that the mOS of the ALKLow group was significantly worse than that of the ALKHigh group [mOS: 4.0 months (95% CI: 2.9-5.1 months) versus 11.0 months (95% CI: 8.3-13.7 months), p = 0.009]. Following covariate adjustment using a Cox regression model, it was indicated that the level of abnormal expression of the ALK protein was an independent prognostic factor for patients with SCLC (HR: 0.486, 95% CI: 0.271-0.871, p = 0.015). Conclusion The prognosis for patients with SCLC with strong abnormal expression of the ALK protein was significantly better than those with weak expression.
Collapse
Affiliation(s)
- Jinhe Xu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wenting Zhang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feilai Xie
- Pathology Department, Dongfang Hospital of Xiamen University, Fuzhou General Hospital of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Chenxi Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Feng Cheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ruiying Rao
- Department of Respiratory and Critical Care Medicine, Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine (900 Hospital), Fuzhou, China
| | - Ying Chen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Wen Wen
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zhongquan Zhao
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Jialing Yuan
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Yuqin Zheng
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zongyang Yu
- Department of Pulmonary and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| |
Collapse
|
12
|
Lim JU, Ryu WK, Park N, Choi J, Lee E, Lee SY, Lim JH. Current and future perspectives in extensive-stage small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359251326705. [PMID: 40093978 PMCID: PMC11909689 DOI: 10.1177/17588359251326705] [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: 11/19/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Small-cell lung cancer (SCLC) is a highly aggressive and rapidly proliferative malignancy that has historically had limited therapeutic advancements. Recent advancements in the understanding of SCLC have led to attempts at subtyping the disease based on transcription factor characteristics, offering new insights into its biology and potential therapeutic targets. In addition, significant progress has been made in developing treatment regimens, providing new hope for improved patient outcomes. The introduction of immune checkpoint inhibitors, such as atezolizumab and durvalumab, in combination with traditional chemotherapy, has marked a significant advancement, demonstrating improved overall survival and progression-free survival compared to chemotherapy alone. Despite these advancements, the prognosis for extensive-stage SCLC (ES-SCLC), the more advanced form of SCLC, remains poor, highlighting the critical need for ongoing research and the development of novel therapeutic strategies. New treatment modalities, such as lurbinectedin and anti-Delta-like Canonical Notch Ligand 3 antibodies, are now included in the treatment options for refractory SCLC, and many more treatment strategies involving combination therapies are being studied. Advances in molecular profiling and the identification of biomarkers are aiding in the development of personalized treatment approaches. This review focuses on these recent advancements and emerging strategies in the treatment of ES-SCLC.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woo Kyung Ryu
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Nuri Park
- Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Juwhan Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eunyoung Lee
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Yun Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, Republic of Korea
- Central R&D Center, Medical & Bio Decision Co., Ltd., Suwon, Republic of Korea
| | - Jun Hyeok Lim
- Center for Lung Cancer, Division of Pulmonology, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, 27, Inhang‑Ro, Jung‑Gu, Incheon 22332, Republic of Korea
| |
Collapse
|
13
|
Watanabe H, Inoue Y, Tsuchiya K, Asada K, Suzuki M, Ogawa H, Tanahashi M, Watanabe T, Matsuura S, Yasuda K, Ohnishi I, Imokawa S, Yasui H, Karayama M, Suzuki Y, Hozumi H, Furuhashi K, Enomoto N, Fujisawa T, Funai K, Shinmura K, Sugimura H, Inui N, Suda T. Lethal co-expression intolerance underlies the mutually exclusive expression of ASCL1 and NEUROD1 in SCLC cells. NPJ Precis Oncol 2025; 9:74. [PMID: 40082639 PMCID: PMC11906894 DOI: 10.1038/s41698-025-00860-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Small cell lung cancer (SCLC) subtypes, defined by the expression of lineage-specific transcription factors (TFs), are thought to be mutually exclusive, with intra-tumoral heterogeneities. This study investigated the mechanism underlying this phenomenon with the aim of identifying a novel vulnerability of SCLC. We profiled the expression status of ASCL1, NEUROD1, POU2F3, and YAP1 in 151 surgically obtained human SCLC samples. On subtyping, a high degree of mutual exclusivity was observed between ASCL1 and NEUROD1 expression at the cell, but not tissue, level. Inducible co-expression models of all combinations of ASCL1, NEUROD1, POU2F3, YAP1, and ATOH1 using SCLC cell lines showed that some expression combinations, such as ASCL1 and NEUROD1, exhibited mutual repression and caused growth inhibition and apoptosis. Gene expression and ATAC-seq analyses of the ASCL1 and NEUROD1 co-expression models revealed that co-expression of ASCL1 in NEUROD1-driven cells, and of NEUROD1 in ASCL1-driven cells, both (although more efficiently by the former) reprogrammed the cell lineage to favor the ectopically expressed factor, with rewiring of chromatin accessibility. Mechanistically, co-expressed NEUROD1 in ASCL1-driven SCLC cells caused apoptosis by downregulating BCL2, likely in a MYC-independent manner. In conclusion, lethal co-expression intolerance underlies the mutual exclusivity between these pioneer TFs, ASCL1 and NEUROD1, in an SCLC cell. Further investigation is warranted to enable therapeutic targeting of this vulnerability.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Kazuo Tsuchiya
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiro Asada
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Makoto Suzuki
- Department of Pathology, Shizuoka General Hospital, Shizuoka, Japan
| | - Hiroshi Ogawa
- Department of Pathology, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Masayuki Tanahashi
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Takuya Watanabe
- Division of Thoracic Surgery, Respiratory Disease Center, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Shun Matsuura
- Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Kazuyo Yasuda
- Department of Pathology, Shizuoka General Hospital, Shizuoka, Japan
- Department of Pathology, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Ippei Ohnishi
- Division of Pathology, Iwata City Hospital, Iwata, Japan
| | - Shiro Imokawa
- Department of Respiratory Medicine, Iwata City Hospital, Iwata, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Chemotherapy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhito Funai
- First Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
14
|
Tang Y, Niu Y, Chen Y, Zhou X, Hu Y, Sun L, Xiong Y, Xu Y, Wang Q, Wang Y, Guo L. Targeting FOXP1 phase separation in small cell lung cancer mechanisms of chemotherapy resistance. Commun Biol 2025; 8:431. [PMID: 40082538 PMCID: PMC11906602 DOI: 10.1038/s42003-025-07804-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
Our study elucidates the role of FOXP1 in chemoresistance in small cell lung cancer(SCLC). FOXP1 enhances chemoresistance by regulating SP8 expression through its super-enhancer (SP8-SE), with SP8 mediating resistance via the homologous recombination repair (HRR) pathway. We also discovered that FOXP1 forms punctate nuclear structures indicative of liquid-liquid phase separation, crucial for its transcriptional regulation. Targeting the FOXP1-SP8-HR axis with BRD4 and PARP inhibitors showed synergistic effects in reducing tumor growth in vitro and in patient-derived xenograft models. These findings identify FOXP1 as a critical mediator and marker of chemoresistance in SCLC, providing a foundation for developing targeted therapies to overcome this resistance.
Collapse
Affiliation(s)
- Yichun Tang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuchun Niu
- Department of Radiation Oncology, The First People's Hospital of Foshan, Cancer Hospital, Foshan, China
| | - Yi Chen
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuyang Zhou
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yueyang Hu
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lei Sun
- Department of Oncology, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, China
| | - Yan Xiong
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yue Xu
- Qingyuan People's Hospital, Qingyuan, China
| | - Qiongyao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yu Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Chen H, Deng C, Gao J, Wang J, Fu F, Wang Y, Wang Q, Zhang M, Zhang S, Fan F, Liu K, Yang B, He Q, Zheng Q, Shen X, Wang J, Hu T, Zhu C, Yang F, He Y, Hu H, Wang J, Li Y, Zhang Y, Cao Z. Integrative spatial analysis reveals tumor heterogeneity and immune colony niche related to clinical outcomes in small cell lung cancer. Cancer Cell 2025; 43:519-536.e5. [PMID: 39983726 DOI: 10.1016/j.ccell.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/12/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Recent advances have shed light on the molecular heterogeneity of small cell lung cancer (SCLC), yet the spatial organizations and cellular interactions in tumor immune microenvironment remain to be elucidated. Here, we employ co-detection by indexing (CODEX) and multi-omics profiling to delineate the spatial landscape for 165 SCLC patients, generating 267 high-dimensional images encompassing over 9.3 million cells. Integrating CODEX and genomic data reveals a multi-positive tumor cell neighborhood within ASCL1+ (SCLC-A) subtype, characterized by high SLFN11 expression and associated with poor prognosis. We further develop a cell colony detection algorithm (ColonyMap) and reveal a spatially assembled immune niche consisting of antitumoral macrophages, CD8+ T cells and natural killer T cells (MT2) which highly correlates with superior survival and predicts improving immunotherapy response in an independent cohort. This study serves as a valuable resource to study SCLC spatial heterogeneity and offers insights into potential patient stratification and personalized treatments.
Collapse
Affiliation(s)
- Haiquan Chen
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Chaoqiang Deng
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Jian Gao
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jun Wang
- School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Fangqiu Fu
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yue Wang
- Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Qiming Wang
- School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Mou Zhang
- School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Shiyue Zhang
- School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Fanfan Fan
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kun Liu
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Bo Yang
- Department of Life and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Qiming He
- Department of Life and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Qiang Zheng
- Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xuxia Shen
- Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jin Wang
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd, Xiamen 361000, China
| | - Tao Hu
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd, Xiamen 361000, China
| | - Changbin Zhu
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd, Xiamen 361000, China
| | - Fei Yang
- Janssen China Research & Development, Shanghai 200233, China
| | - Yonghong He
- Department of Life and Health, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Hong Hu
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jialei Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yuan Li
- Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yang Zhang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Institute of Thoracic Oncology, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhiwei Cao
- School of Life Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Chen Y, Liu X, Sun Y, Liu K, Ding D, Song S, Tan W. Noninvasive molecular imaging using anti-Trop-2 aptamer for targeted therapy of small cell lung cancer. J Nanobiotechnology 2025; 23:182. [PMID: 40050871 PMCID: PMC11887224 DOI: 10.1186/s12951-025-03184-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/01/2025] [Indexed: 03/09/2025] Open
Abstract
Recent advancements in antibody-drug conjugates (ADCs) targeting trophoblast surface cell antigen 2 (Trop-2) have brought important progress in the field of targeted therapy. This progress also holds promise for the treatment of small cell lung cancer (SCLC) as anti-Trop-2 therapy appears to have a safe and effective clinical activity in metastatic SCLC patients. However, effective treatments of anti-Trop-2 ADCs rely on the comprehensive assessment of Trop-2 expression at the tumor sites, SCLC exhibits intratumoral heterogeneity, making the accurate acquisition of histological biopsies a challenge. To address this issue, we herein report the development of an anti-Trop-2 aptamer consisting of 76 bases is specifically bind to Trop-2-overexpressing SCLC cells. Further truncated anti-Trop-2 aptamer with 46 nucleotides also possesses excellent in vitro and in vivo binding affinity with Trop-2 antigens. After radiolabeling with gallium-68 radionuclide, an aptamer-based molecular imaging probe was successfully fabricated named [68Ga]Ga-NOTA-TRP-c. This imaging probe demonstrated effective and precise differentiation of Trop-2-positive tumors in both murine- and human-derived animal models, exhibiting favorable metabolic profiles. Furthermore, Trop-2-positive SCLC tumors recognized by anti-Trop-2 aptamer can be treated with anti-Trop-2 ADC sacituzumab govitecan (SG), either in vitro or in vivo. Importantly, SG induces DNA damage and cell apoptosis without affecting the expression of Trop-2 on the cell surface, which makes it possible to use anti-Trop-2 aptamer to monitor the expression of Trop-2 in SCLC. This study highlights the potential of aptamer-based molecular imaging and imaging-guided SG treatment as a promising option for targeted therapy in SCLC.
Collapse
Affiliation(s)
- Yamei Chen
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Xuwei Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Keying Liu
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
17
|
Pérez-Cabello JA, Artero-Castro A, Molina-Pinelo S. Small cell lung cancer unveiled: Exploring the untapped resource of circulating tumor cells-derived organoids. Crit Rev Oncol Hematol 2025; 207:104622. [PMID: 39832682 DOI: 10.1016/j.critrevonc.2025.104622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
Small cell lung cancer (SCLC) remains a challenge in oncology due to its aggressive behavior and dismal prognosis. Despite advances in treatments, novel strategies are urgently needed. Enter liquid biopsy-a game-changer in SCLC management. This revolutionary non-invasive approach allows for the analysis of circulating tumor cells (CTCs), offering insights into tumor behavior and treatment responses. Our review focuses on a groundbreaking frontier: harnessing CTCs to create three-dimensional (3D) organoid models. These models, derived from CTCs that break away from the primary tumor or metastatic locations, hold immense potential for revolutionizing cancer research, especially in SCLC. We explore the essential conditions for successfully establishing CTC-derived organoids-a transformative approach with profound implications for personalized medicine. Our evaluation spans diverse isolation techniques, shedding light on their advantages and limitations. Furthermore, we uncover the critical factors governing the cultivation of 3D organoids from CTCs, meticulously mimicking the tumor microenvironment. This review comprehensively elucidates the molecular characterization of these organoids, showcasing their potential in identifying treatment targets and predicting responses. In essence, our review amalgamates cutting-edge methodologies for isolating CTCs, establishing transformative CTC-derived organoids, and characterizing their molecular landscape. This represents a promising frontier for advancing personalized medicine in the complex realm of SCLC management and holds significant implications for translational research.
Collapse
Affiliation(s)
- Jesús A Pérez-Cabello
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain
| | - Ana Artero-Castro
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain
| | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, University of Seville, Seville 41013, Spain; Spanish Center for Biomedical Research Network in Oncology (CIBERONC), Madrid 28029, Spain.
| |
Collapse
|
18
|
Zhu Y, Wu J, Wang H, Chi K, Diao X, Zhuo M, Lin D. Whole-section digital analysis of immune profiles in surgically resected small cell lung carcinoma and their associations with molecular subtypes. Transl Lung Cancer Res 2025; 14:449-466. [PMID: 40114934 PMCID: PMC11921296 DOI: 10.21037/tlcr-24-924] [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: 10/08/2024] [Accepted: 01/09/2025] [Indexed: 03/22/2025]
Abstract
Background The molecular subtype-specific features of the tumor immune microenvironment (TIME) in small cell lung carcinoma (SCLC) remain poorly understood. We aimed to analyze the immune profiles in surgically resected SCLC and their associations with molecular subtypes. Methods Tumor samples from 83 treatment-naive SCLC patients who underwent surgical resection were analyzed. The protein expression of subtype-defining markers (ASCL1, NEUROD1, POU2F3, and YAP1) and nine immune-related markers were assessed using whole-section immunohistochemistry. Digital image analysis was employed for precise quantification of immune cell infiltrates and distributions. The findings were subsequently correlated with clinicopathological parameters and patient prognoses. Results Unsupervised hierarchical clustering categorized the tumors into four molecular subtypes: achaete-scute homologue 1-dominant (ASCL1; SCLC-A, 71.1%, n=59), neuronal differentiation factor 1-dominant (NEUROD1; SCLC-N, 12.1%, n=10), POU class 2 homeobox 3-dominant (POU2F3; SCLC-P, 10.8%, n=9), and quadruple-negative (SCLC-QN, 6.0%, n=5). Expression of major histocompatibility complex class I (MHC I) and class II (MHC II; P=0.02, P=0.02), tumor programmed death-ligand 1 (PD-L1; P=0.006), and an inflamed phenotype characterized by CD8+/CD3+ T cells (P=0.001, P=0.003) were more prominent in SCLC-P tumors compared to other subtypes. Additionally, SCLC-P tumors demonstrated the highest levels of MHC II (P=0.04) and PD-L1 expression on both tumor and stromal cells (P=0.003, P=0.01). The tumor proportion score of PD-L1 positively correlated with tumor expression levels of POU2F3 (rho=0.297, P=0.006) and MHC I (rho=0.239, P=0.03), as well as the combined positive score of PD-L1 (rho=0.222, P=0.04; rho=0.433, P<0.001). Intra-tumoral tertiary lymphoid structures (intra-TLS) and peri-tumoral TLS (peri-TLS) were observed in 60.2% (n=50) and 96.4% (n=80) of patients, respectively. High intra-TLS density was more frequently associated with SCLC-P tumors (P=0.02). Notably, low peri-TLS density and stromal PD-L1 expression were linked to improved overall survival (OS) and progression-free survival (PFS), respectively. Conclusions This study highlights the heterogeneity of the TIME across molecular subtypes of SCLC. The SCLC-P subtype and MHC I expression may serve as predictive biomarkers for immunotherapy response, while peri-TLS density and stromal PD-L1 expression might serve as prognostic indicators in resected SCLC.
Collapse
Affiliation(s)
- Yanli Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianghua Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Haiyue Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaiwen Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinting Diao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
19
|
Gao R, Wu P, Yin X, Zhuang L, Meng X. Deep analysis of the trials and major challenges in the first-line treatment for patients with extensive-stage small cell lung cancer. Int Immunopharmacol 2025; 148:114116. [PMID: 39847950 DOI: 10.1016/j.intimp.2025.114116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The median overall survival (OS) is approximately 10 months when chemotherapy alone is the first-line treatment for extensive-stage small cell lung cancer (ES-SCLC). The approval of the two PD-L1 inhibitors, atezolizumab and durvalumab, marked the beginning of the immunotherapy era for ES-SCLC. Serplulimab, as the first PD-1 inhibitor to achieve success in the first-line treatment of ES-SCLC, has not only demonstrated significant improvements in patient survival outcomes but also ushered in a new era for PD-1 inhibitors in the treatment of ES-SCLC. Recently, antiangiogenic agents with chemo-immunotherapy have achieved breakthroughs in first-line ES-SCLC treatment. Improving the clinical benefits of individualized treatment for patients with ES-SCLC remains challenging. Challenges include identifying biomarkers for targeted therapy, exploring new treatments, developing new medicines, and classifying SCLC molecular subtypes. This review provides an in-depth analysis of research on first-line ES-SCLC treatment. Additionally, it discusses advances in ES-SCLC treatment.
Collapse
Affiliation(s)
- Ran Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Peizhu Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Xiaoyan Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China
| | - Lulu Zhuang
- Cheeloo College of Cancer Center, Shandong University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, Jinan, China.
| |
Collapse
|
20
|
Chen YQ, Tan JX, Gao LL, Yang JX, Huang J, Yang JJ, Zhao Q. Exploring YAP1-related TIME in SCLC: implications for survival and treatment response to immuno-chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:8. [PMID: 40051494 PMCID: PMC11883233 DOI: 10.20517/cdr.2024.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/30/2024] [Accepted: 01/25/2025] [Indexed: 03/09/2025]
Abstract
Aim: Small-cell lung cancer (SCLC) is usually diagnosed as an advanced stage with a poor outcome. SCLC has limited response to immunotherapy due to the absence or lack of immune cell infiltration, so studying its tumor immune microenvironment (TIME) is essential. Methods: The study involved patients with extensive-stage small-cell lung cancer (ES-SCLC) diagnosed at the Guangdong Lung Cancer Institute between January 2018 and April 2022 who had received the atezolizumab/carboplatin/etoposide (ECT) treatment. We used multi-immunohistochemistry (mIHC) to assess the prognostic value of YAP1 and TIME in SCLC, with results confirmed using public data. Results: 15 patients with sufficient baseline biopsy samples were included in this study. The total population of YAP1-positive cells is inversely related to progression-free survival (PFS) and shows a potential negative correlation with overall survival (OS). CD56-positive cells are the primary components of TIME in SCLC tumor parenchyma and stroma. The total population and cell density of YAP1-positive cells are significantly positively correlated with CD4-positive cells. Furthermore, in the tumor parenchyma, both the proportion and the cell density of YAP1-positive cells are positively correlated with that of FOXP3-positive cells. The total population of CD56-positive cells showed a negative correlation trend with YAP1-positive cells but without significant difference. Conclusion: YAP1 has shown prognostic value in SCLC patients receiving ECT regimen treatment. The high expression level of YAP1 seems related to the inhibitory TIME. However, some prospective studies with larger populations are warranted.
Collapse
Affiliation(s)
- Yu-Qing Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Authors contributed equally
| | - Jia-Xiong Tan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Authors contributed equally
| | - Ling-Ling Gao
- Beihang University, Beijing 100191, China
- Authors contributed equally
| | - Jia-Xing Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Qiang Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
21
|
Morin A, Chu CP, Pavlidis P. Identifying Reproducible Transcription Regulator Coexpression Patterns with Single Cell Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.15.580581. [PMID: 38559016 PMCID: PMC10979919 DOI: 10.1101/2024.02.15.580581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The proliferation of single cell transcriptomics has potentiated our ability to unveil patterns that reflect dynamic cellular processes such as the regulation of gene transcription. In this study, we leverage a broad collection of single cell RNA-seq data to identify the gene partners whose expression is most coordinated with each human and mouse transcription regulator (TR). We assembled 120 human and 103 mouse scRNA-seq datasets from the literature (>28 million cells), constructing a single cell coexpression network for each. We aimed to understand the consistency of TR coexpression profiles across a broad sampling of biological contexts, rather than examine the preservation of context-specific signals. Our workflow therefore explicitly prioritizes the patterns that are most reproducible across cell types. Towards this goal, we characterize the similarity of each TR's coexpression within and across species. We create single cell coexpression rankings for each TR, demonstrating that this aggregated information recovers literature curated targets on par with ChIP-seq data. We then combine the coexpression and ChIP-seq information to identify candidate regulatory interactions supported across methods and species. Finally, we highlight interactions for the important neural TR ASCL1 to demonstrate how our compiled information can be adopted for community use.
Collapse
Affiliation(s)
- Alexander Morin
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, BC, Canada
| | - C. Pan Chu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, BC, Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Zhu Y, Ren W, Li S, Wu J, Hu X, Wang H, Chi K, Zhuo M, Lin D. Heterogeneity of molecular subtyping and therapy-related marker expression in primary tumors and paired lymph node metastases of small cell lung cancer. Virchows Arch 2025; 486:243-255. [PMID: 38347268 DOI: 10.1007/s00428-024-03754-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/18/2024] [Accepted: 01/27/2024] [Indexed: 03/04/2025]
Abstract
The classification of molecular subtypes and the identification of targetable molecules have been proposed for small cell lung cancer (SCLC) patients. Our aim was to investigate whether the expression of these markers evaluated using lymph node (LN) metastases represents that of primary tumors. We enrolled 46 surgically resected SCLC patients' primary tumors and paired mediastinal LN metastases. The protein expression of subtype-defining markers (ASCL1, NEUROD1, POU2F3, and YAP1) and therapeutic markers (DLL3, MYC, PD-L1, and MHC I) was examined by immunohistochemistry and was correlated with clinicopathological parameters and prognoses. In primary and metastatic tumors, the expression of these markers was 78.3% and 87.0%, 50.0% and 63.0%, 13.0% and 6.5%, 17.4% and 15.2%, 84.8% and 87.0%, 17.4% and 6.5%, 50.0% and 34.8%, and 60.9% and 37.0%, respectively. Positive tumor PD-L1 expression was less present in LN metastases (p = 0.015), and the same was true for MHC I expression (p = 0.036). NEUROD1 and DLL3 expression levels in metastatic tumors were stronger (p < 0.001 and p = 0.002, respectively); conversely, POU2F3, MYC, PD-L1, and MHC I expression levels were weaker (p = 0.018, p = 0.019, p = 0.001, and p < 0.001, respectively). In 15 (32.6%) patients, we observed a change in the molecular subtyping pattern, and a higher number of neuroendocrine (NE)-high phenotype patients were diagnosed when using the LN specimens (91.3% vs. 84.8%). TNM stage and postoperative chemotherapy were independent prognostic factors in surgically resected SCLC patients, and no prognostic differences were found among molecular subtypes. This study highlights the discordance of subtype-specific proteins and therapeutic markers between SCLC primary tumors and LN metastases. Additionally, our findings have therapeutic and prognostic implications and warrant further clinical investigation.
Collapse
Affiliation(s)
- Yanli Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Wenhao Ren
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Sheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianghua Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiao Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Haiyue Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaiwen Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
23
|
Yuan L, Li S, Zhu Y, Yang L, Zhang X, Qu Y, Wang Z, Duan J, Zhong J, Tian Y, Liu L, Sun B, Fei K, Liu Z, Zhang J, He Y, Guo Y, He D, Zhuang W, Zhang J, Ma Z, Bai H, Wang J. ATAD2 is a potential immunotherapy target for patients with small cell lung cancer harboring HLA-A∗0201. EBioMedicine 2025; 112:105515. [PMID: 39808946 PMCID: PMC11782892 DOI: 10.1016/j.ebiom.2024.105515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) represents a highly aggressive neuroendocrine tumour with a dismal prognosis. Currently, the identification of a specific tumour antigen that can facilitate immune-based therapies for SCLC remains elusive. METHODS We employed liquid chromatography-tandem mass spectrometry (LC-MS/MS) to analyse cancer/testis antigens (CTAs) in SCLC cell lines and human tumour specimens. Immunohistochemistry of clinical specimens was performed to compare protein expression in SCLC, non-small cell lung cancer (NSCLC), and matched normal-adjacent tissues. Additionally, publicly available RNA sequencing databases were interrogated to identify gene expression patterns in different SCLC subtypes and in different disease stages. FINDINGS Distinct numbers and types of CTAs were identified across SCLC subtypes, with significantly higher expression levels of ATPase family AAA domain-containing protein 2 (ATAD2) observed in SCLC compared to normal adjacent tissues and NSCLC tissues. A dynamic expression pattern of ATAD2 was found throughout the clinical course of SCLC and exhibited a positive correlation with achaete-scute family bHLH transcription factor 1 (ASCL1) expression in SCLC. Immunopeptidomics analysis identified the YSDDDVPSV sequence derived from the HLA-A∗02:01 restriction epitope of ATAD2 as a highly promising tumour antigen candidate for potential immunotherapy applications. YSDDDVPSV immunopeptides were confirmed to be present in SCLC-A and SCLC-N with HLA-A∗02:01 restriction. Notably, HLA-A∗02:01 T cells exhibited a robust response upon stimulation with YSDDDVPSV immunopeptide pulsed by T2 cells. INTERPRETATION Our findings highlight the potential of targeting the ATAD2 YSDDDVPSV immunopeptide for SCLC immunotherapy, thereby offering a promising avenue for the development of adoptive T cell therapies to effectively treat ASCL1-positive or NEUROD1-positive SCLC carrying HLA-A∗02:01. FUNDING This study was supported by the National key R&D program of China (2022YFC2505000); National Natural Science Foundation of China (NSFC) general program (82272796) NSFC special program (82241229); CAMS Innovation Fund for Medical Sciences (CIFMS 2022-I2M-1-009); CAMS Key Laboratory of Translational Research on Lung Cancer (2018PT31035); Aiyou foundation (KY201701). National key R&D program of China (2022YFC2505004). NSFC general program (81972905). Medical Oncology Key Foundation of Cancer Hospital Chinese Academy of Medical Sciences (CICAMS-MOCP2022012).
Collapse
Affiliation(s)
- Li Yuan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Sini Li
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yixiang Zhu
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Lin Yang
- Department of Pathology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xue Zhang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Yan Qu
- Department of Radiotherapy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Jia Zhong
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Yanhua Tian
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Lihui Liu
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Boyang Sun
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Kailun Fei
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Zheng Liu
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Jian Zhang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Yan He
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Yufeng Guo
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - DanMing He
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Wei Zhuang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Jinsong Zhang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Zixiao Ma
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
24
|
Meador CB, Digumarthy SR, Yeap BY, Hung YP, Mino-Kenudson M, Farago AF, Heist RS, Marcoux JP, Rangachari D, Barbie DA, Piotrowska Z. Phase I/II Investigator-Initiated Study of Olaparib and Temozolomide in SCLC: Final Analysis and CNS Outcomes. Clin Cancer Res 2025; 31:25-34. [PMID: 39470668 DOI: 10.1158/1078-0432.ccr-24-2350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/12/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Temozolomide plus PARP inhibition has shown promise in small cell lung cancer (SCLC). We previously reported outcomes from the first 50 patients (cohort 1) of a phase I/II trial of olaparib/temozolomide in recurrent SCLC. In this study, we report a final analysis of this trial, including a second cohort with an alternate dosing strategy and an exploratory analysis of central nervous system (CNS)-specific outcomes. PATIENTS AND METHODS This was an open-label phase I/II trial testing the combination of olaparib and temozolomide in relapsed SCLC. The primary endpoint was objective response rate (ORR). Secondary endpoints were safety, progression-free survival, and overall survival. We tested escalating doses of olaparib/temozolomide across two cohorts, both of which had temozolomide dosed on days 1 to 7 of each 21-day cycle. In previously published cohort 1, olaparib was dosed on days 1 to 7; in cohort 2, olaparib was dosed continuously. RESULTS Sixty-six patients were enrolled across the two cohorts: 50 in cohort 1 and 16 in cohort 2. The confirmed ORR of cohort 1 was 41.7% (20/48 evaluable), and the confirmed ORR of cohort 2 was 7% (1/14 evaluable; closed after dose escalation to enrollment for lack of observed efficacy). Among 15/66 patients (22.7%) with untreated brain metastases at enrollment, the best overall intracranial response was complete response in 6/15 patients, partial response in 4/15 patients, and stable disease in 3/15 patients for a CNS disease control rate of 87% (95% confidence interval, 59.5%-98.3%). CONCLUSIONS Olaparib/temozolomide may be effective in relapsed SCLC, especially for patients with CNS disease. Ongoing analyses with regard to optimal dosing schedule will inform potential for future use of this combination in SCLC.
Collapse
Affiliation(s)
- Catherine B Meador
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Subba R Digumarthy
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Beow Y Yeap
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Anna F Farago
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Rebecca S Heist
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | | | - Deepa Rangachari
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Zofia Piotrowska
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
25
|
Sztankovics D, Szalai F, Moldvai D, Dankó T, Scheich B, Pápay J, Sebestyén A, Krencz I. Comparison of molecular subtype composition between independent sets of primary and brain metastatic small cell lung carcinoma and matched samples. Lung Cancer 2025; 199:108071. [PMID: 39721126 DOI: 10.1016/j.lungcan.2024.108071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Recent advances in the subclassification of small cell lung carcinomas (SCLCs) may help to overcome the unmet need for targeted therapies and improve survival. However, limited information is available on how the expression of the subtype markers changes during tumour progression. Our study aimed to compare the expression of these markers in primary and brain metastatic SCLCs. MATERIALS AND METHODS Immunohistochemical analysis of the subtype markers was performed on 120 SCLCs (including 10 matched samples) and SCLC xenografts. RESULTS Compared to primary SCLCs, there was a significant increase in the proportion of mixed subtypes in brain metastases, with a rate of ASCL1high/NeuroD1high and ASCL1high/NeuroD1high/YAP1high subtypes increasing to 48 % and 18 %, respectively. The subtype of the paired samples matched in only one-third of the cases. Although we did not observe a significant change after chemotherapy, a continuous decrease in ASCL1 expression coupled with an increase in the NeuroD1 expression was detected in the xenografts in a long-term experiment. DISCUSSION Our results indicate that the expression of subtype markers frequently changes during disease progression, and subtype analysis of the primary SCLC may not provide accurate information about the characteristics of the recurrent or metastatic tumour. Therefore, repeated sampling and subtyping may be necessary for subtype-specific targeted therapy.
Collapse
Affiliation(s)
- Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Titanilla Dankó
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői, út 26., H-1085 Budapest, Hungary.
| |
Collapse
|
26
|
Zhu Y, Li S, Wang H, Ren W, Chi K, Wu J, Mao L, Huang X, Zhuo M, Lin D. Molecular subtypes, predictive markers and prognosis in small-cell lung carcinoma. J Clin Pathol 2024; 78:42-50. [PMID: 37775262 DOI: 10.1136/jcp-2023-209109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/14/2023] [Indexed: 10/01/2023]
Abstract
AIMS A new molecular subtype classification was proposed for small-cell lung carcinoma (SCLC). We aimed to further validate the classification in various SCLC patient samples using immunohistochemistry (IHC) to highlight its clinical significance. METHODS We analysed the protein expression of four subtype (achaete-scute family BHLH transcription factor 1 (ASCL1), neuronal differentiation 1 (NEUROD1), POU class 2 homeobox 3 (POU2F3) and Yes1-associated transcriptional regulator (YAP1)) and two predictive markers (delta-like ligand 3 (DLL3) and MYC) using IHC in 216 specimens from 195 SCLC patients, including 21 pairs of resected biopsy tumours. Associations among molecular subtypes, clinicopathological features and prognostic implications were also explored. RESULTS The ASCL1, NEUROD1, POU2F3, YAP1, DLL3 and MYC-positive expression rates were 70.3%, 56.9%, 14.9%, 19.0%, 75.4% and 22.6%, respectively. DLL3 expression had positive and negative associations with that of ASCL1 and POU2F3/YAP1, respectively, whereas MYC had the opposite effect. Strong associations of ASCL1 (Ρ=0.8603, p<0.0001), NEUROD1 (Ρ=0.8326, p<0.0001), POU2F3 (Ρ=0.6950, p<0.0001) and YAP1 (Ρ=0.7466, p<0.0001) expressions were detected between paired resected biopsy tumours. In addition to SCLC-A (ASCL1-dominant), SCLC-N (NEUROD1-dominant) and SCLC-P (POU2F3-dominant), unsupervised hierarchical cluster analyses identified a fourth, quadruple-negative SCLC subtype (SCLC-QN) characterised by the low expression of all four subtype-specific proteins, and 55.4% (n=108), 27.2% (n=53), 11.8% (n=23) and 5.6% (n=11) were categorised as SCLC-A, SCLC-N, SCLC-P and SCLC-QN, respectively. Significant enrichment of SCLC-P in the combined SCLC cohort was observed, and adenocarcinoma was more prevalent in SCLC-A, while large-cell neuroendocrine carcinoma was more commonly seen in SCLC-P. No survival difference was found among molecular subtypes. CONCLUSIONS Our results provide clinical insights into the diagnostic, prognostic and predictive significance of SCLC molecular subtype classifications.
Collapse
Affiliation(s)
- Yanli Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Sheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Haiyue Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Wenhao Ren
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaiwen Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianghua Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Luning Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaozheng Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department I of Thoracic Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
27
|
Yu Z, Zou J, Xu F. The molecular subtypes of small cell lung cancer defined by key transcription factors and their clinical significance. Lung Cancer 2024; 198:108033. [PMID: 39571251 DOI: 10.1016/j.lungcan.2024.108033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Lung cancer, a prevalent and deadly malignancy, is classified into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). SCLC is further subdivided into four molecular subtypes-SCLC-A, SCLC-N, SCLC-P, and SCLC-I-based on key transcription factor expression. METHODS Immunohistochemistry (IHC) was used to assess ASCL1, NEUROD1, and POU2F3 expression in tumor tissues. The H-Score quantified these results. Clinical characteristics, overall survival (OS), progression-free survival (PFS), and treatment responses were analyzed by subtype, and sensitivity to different treatments was assessed. Risk factors were identified through univariate and multivariate analyses. RESULTS IHC and H-Score analysis showed that POU2F3 expression was mutually exclusive with ASCL1 or NEUROD1. Subtype distribution was as follows: SCLC-A (40 %), SCLC-N (33 %), SCLC-P (7 %), and SCLC-I (20 %). There were no significant differences in baseline characteristics, OS (p = 0.829), or PFS (p = 0.924) among subtypes. However, the SCLC-I subtype showed a trend toward improved outcomes with platinum-based doublet chemotherapy plus immune checkpoint inhibitors. Multivariate COX regression identified M stage (HR: 1.72, 95 % CI: 1.13-2.63, p = 0.012) and bone metastasis at diagnosis (HR: 1.58, 95 % CI: 1.02-2.43, p = 0.040) as independent risk factors. CONCLUSION This study confirmed the SCLC subtyping based on key transcription factors. While no significant differences in OS and PFS among subtypes were found, the SCLC-I subtype showed potential benefit from platinum-based chemotherapy combined with immune checkpoint inhibitors. M stage and bone metastasis at diagnosis were identified as independent risk factors for SCLC.
Collapse
Affiliation(s)
- Zhuchen Yu
- Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Juntao Zou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| | - Fei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
| |
Collapse
|
28
|
Miglietta G, Russo M, Capranico G, Marinello J. Stimulation of cGAS-STING pathway as a challenge in the treatment of small cell lung cancer: a feasible strategy? Br J Cancer 2024; 131:1567-1575. [PMID: 39215193 PMCID: PMC11555062 DOI: 10.1038/s41416-024-02821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Lung cancer has a significant incidence among the population and, unfortunately, has an unfavourable prognosis in most cases. The World Health Organization (WHO) classifies lung tumours into two subtypes based on their phenotype: the Non-Small Cell Lung Cancer (NSCLC) and the Small Cell Lung Cancer (SCLC). SCLC treatment, despite advances in chemotherapy and radiotherapy, is often unsuccessful for cancer recurrence highlighting the need to develop novel therapeutic strategies. In this review, we describe the genetic landscape and tumour microenvironment that characterize the pathological processes of SCLC and how they are responsible for tumour immune evasion. The immunosuppressive mechanisms engaged in SCLC are critical factors to understand the failure of immunotherapy in SCLC and, conversely, suggest that new signalling pathways, such as cGAS/STING, should be investigated as possible targets to stimulate an innate immune response in this subtype of lung cancer. The full comprehension of the innate immunity of cancer cells is thus crucial to open new challenges for successful immunotherapy in treating SCLC and improving patient outcomes.
Collapse
Affiliation(s)
- Giulia Miglietta
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Marco Russo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giovanni Capranico
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| | - Jessica Marinello
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
| |
Collapse
|
29
|
Ozakinci H, Alontaga AY, Cano P, Koomen JM, Perez BA, Beg AA, Chiappori AA, Haura EB, Boyle TA. Unveiling the Molecular Features of SCLC With a Clinical RNA Expression Panel. JTO Clin Res Rep 2024; 5:100723. [PMID: 39386315 PMCID: PMC11459576 DOI: 10.1016/j.jtocrr.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/26/2024] [Accepted: 08/23/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The translation of gene expression profiles of SCLC to clinical testing remains relatively unexplored. In this study, gene expression variations in SCLC were evaluated to identify potential biomarkers. Methods RNA expression profiling was performed on 44 tumor samples from 35 patients diagnosed with SCLC using the clinically validated RNA Salah Targeted Expression Panel (RNA STEP). RNA sequencing (RNA-Seq) and immunohistochemistry were performed on two different SCLC cohorts, and correlation analyses were performed for the ASCL1, NEUROD1, POU2F3, and YAP1 genes and their corresponding proteins. RNA STEP and RNA-Seq results were evaluated for gene expression profiles and heterogeneity between SCLC primary and metastatic sites. RNA STEP gene expression profiles of independent SCLC samples (n = 35) were compared with lung adenocarcinoma (n = 160) and squamous cell carcinoma results (n = 25). Results The RNA STEP results were highly correlated with RNA-Seq and immunohistochemistry results. The dominant transcription regulator by RNA STEP was ASCL1 in 74.2% of the samples, NEUROD1 in 20%, and POU2F3 in 2.9%. The ASCL1, NEUROD1, and POU2F3 gene expression profiles were heterogeneous between primary and metastatic sites. SCLCs displayed markedly high expression for targetable genes DLL3, EZH2, TERT, and RET. SCLCs were found to have relatively colder immune profiles than lung adenocarcinomas and squamous cell carcinomas, characterized by lower expression of HLA genes, immune cell, and immune checkpoint genes, except the LAG3 gene. Conclusions Clinical-grade SCLC RNA expression profiling has value for SCLC subtyping, design of clinical trials, and identification of patients for trials and potential targeted therapy.
Collapse
Affiliation(s)
- Hilal Ozakinci
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Aileen Y. Alontaga
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Pedro Cano
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John M. Koomen
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Bradford A. Perez
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Florida Cancer Specialists & Research Institute, Trinity Cancer Center, Trinity, Florida
| | - Amer A. Beg
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Alberto A. Chiappori
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Eric B. Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Theresa A. Boyle
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| |
Collapse
|
30
|
Jiang J, Han D, Wang J, Wen W, Zhang R, Qin W. Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e761. [PMID: 39372390 PMCID: PMC11450264 DOI: 10.1002/mco2.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/08/2024] Open
Abstract
Neuroendocrine transdifferentiation (NEtD), also commonly referred to as lineage plasticity, emerges as an acquired resistance mechanism to molecular targeted therapies in multiple cancer types, predominately occurs in metastatic epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors and metastatic castration-resistant prostate cancer treated with androgen receptor targeting therapies. NEtD tumors are the lethal cancer histologic subtype with unfavorable prognosis and limited treatment. A comprehensive understanding of molecular mechanism underlying targeted-induced plasticity could greatly facilitate the development of novel therapies. In the past few years, increasingly elegant studies indicated that NEtD tumors share key the convergent genomic and phenotypic characteristics irrespective of their site of origin, but also embrace distinct change and function of molecular mechanisms. In this review, we provide a comprehensive overview of the current understanding of molecular mechanism in regulating the NEtD, including genetic alterations, DNA methylation, histone modifications, dysregulated noncoding RNA, lineage-specific transcription factors regulation, and other proteomic alterations. We also provide the current management of targeted therapies in clinical and preclinical practice.
Collapse
Affiliation(s)
- Jun Jiang
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
- Department of Health Service, Base of Health ServiceAir Force Medical UniversityXi'anChina
| | - Donghui Han
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| | - Jiawei Wang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, and National Translational Science Center for Molecular MedicineAir Force Medical UniversityXi'anChina
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical ResearchNorthwestern Polytechnical UniversityXi'anChina
| | - Rui Zhang
- State Key Laboratory of Cancer BiologyDepartment of ImmunologyAir Force Medical UniversityXi'anChina
| | - Weijun Qin
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
31
|
Redin E, Quintanal-Villalonga Á, Rudin CM. Small cell lung cancer profiling: an updated synthesis of subtypes, vulnerabilities, and plasticity. Trends Cancer 2024; 10:935-946. [PMID: 39164163 DOI: 10.1016/j.trecan.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with high proliferative and metastatic capacity. SCLC has been classified into molecular subtypes based on differential expression of lineage-defining transcription factors. Recent studies have proposed new subtypes that are based on both tumor-intrinsic and -extrinsic factors. SCLC demonstrates substantial intratumoral subtype heterogeneity characterized by highly plastic transcriptional states, indicating that the initially dominant subtype can shift during disease progression and in association with resistance to therapy. Strategies to promote or constrain plasticity and cell fate transitions have nominated novel targets that could prompt the development of more durably effective therapies for patients with SCLC. In this review, we describe the latest advances in SCLC subtype classification and their biological and clinical implications.
Collapse
Affiliation(s)
- Esther Redin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Álvaro Quintanal-Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology Program, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
32
|
Liu T, Chen X, Mo S, Zhou T, Ma W, Chen G, Chen X, Shi M, Yang Y, Huang Y, Zhao H, Fang W, Yang Y, Li J, Zhang L, Zhao Y. Molecular subtypes and prognostic factors of lung large cell neuroendocrine carcinoma. Transl Lung Cancer Res 2024; 13:2222-2235. [PMID: 39430332 PMCID: PMC11484736 DOI: 10.21037/tlcr-24-292] [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: 04/04/2024] [Accepted: 07/26/2024] [Indexed: 10/22/2024]
Abstract
Background Lung large cell neuroendocrine carcinoma (LCNEC) is an aggressive disease with poor prognosis and short-term survival, which lacks effective prognostic indicators. The study aims to investigate the molecular subtypes and prognostic markers of lung LCNEC. Methods Patients diagnosed with lung LCNEC at Sun Yat-sen University Cancer Center (SYSUCC) between November 2007 and January 2021 were screened. Baseline clinical data were collected and routine blood indexes including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. Immunohistochemistry (IHC) of ASCL1, NEUROD1, POU2F3, YAP1 were done to perform molecular subtyping, while CD56, Syn, CgA, CD3, CD8, CD20, CD68, and CD163 were also stained on tissue samples. Then prognostic factors of lung LCNEC were explored. Results One hundred and fifty-one lung LCNEC patients were identified, 103 of whom had complete clinical information, available routine blood and biochemical indexes were eventually included in the present study. Tumor tissue specimens were available from 64 patients. Positive expression rates of ASCL1, NEUROD1, and YAP1 were 82.8%, 50.0%, and 28.1%, respectively. No POU2F3+ cases were detected. Forty (62.5%) patients co-expressed with two or three markers. High LMR (>3.3) was an independent predictor of favorable prognosis of disease-free survival (DFS) [hazard ratio (HR), 0.391; 95% confidence interval (CI): 0.161-0.948; P=0.04] and overall survival (OS) (HR, 0.201; 95% CI: 0.071-0.574; P=0.003). Notably, high LMR was correlated with higher intra-tumoral CD3+ (P=0.004), CD8+ (P=0.01), and CD68+ (P<0.001) immune cell infiltration compared to low LMR in lung LCNEC. Conclusions Our study validated molecular subtypes by IHC in lung LCNEC, and co-expression was found among different subtypes, with no prognostic effect. High blood LMR level was associated with a favorable prognosis in lung LCNEC, which might partly reflect a hot tumor tissue immune microenvironment. Our findings may benefit clinical practice, and further studies are warranted.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xueyuan Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Silang Mo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjuan Ma
- Department of Intensive Care Unit, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengting Shi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuwen Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongyun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
33
|
Jin Y, Wu Y, Reuben A, Zhu L, Gay CM, Wu Q, Zhou X, Mo H, Zheng Q, Ren J, Fang Z, Peng T, Wang N, Ma L, Fan Y, Song H, Zhang J, Chen M. Single-cell and spatial proteo-transcriptomic profiling reveals immune infiltration heterogeneity associated with neuroendocrine features in small cell lung cancer. Cell Discov 2024; 10:93. [PMID: 39231924 PMCID: PMC11375181 DOI: 10.1038/s41421-024-00703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/23/2024] [Indexed: 09/06/2024] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive pulmonary neuroendocrine malignancy featured by cold tumor immune microenvironment (TIME), limited benefit from immunotherapy, and poor survival. The spatial heterogeneity of TIME significantly associated with anti-tumor immunity has not been systemically studied in SCLC. We performed ultra-high-plex Digital Spatial Profiling on 132 tissue microarray cores from 44 treatment-naive limited-stage SCLC tumors. Incorporating single-cell RNA-sequencing data from a local cohort and published SCLC data, we established a spatial proteo-transcriptomic landscape covering over 18,000 genes and 60 key immuno-oncology proteins that participate in signaling pathways affecting tumorigenesis, immune regulation, and cancer metabolism across 3 pathologically defined spatial compartments (pan-CK-positive tumor nest; CD45/CD3-positive tumor stroma; para-tumor). Our study depicted the spatial transcriptomic and proteomic TIME architecture of SCLC, indicating clear intra-tumor heterogeneity dictated via canonical neuroendocrine subtyping markers; revealed the enrichment of innate immune cells and functionally impaired B cells in tumor nest and suggested potentially important immunoregulatory roles of monocytes/macrophages. We identified RE1 silencing factor (REST) as a potential biomarker for SCLC associated with low neuroendocrine features, more active anti-tumor immunity, and prolonged survival.
Collapse
Affiliation(s)
- Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Yuefeng Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingzhe Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xintong Zhou
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haomin Mo
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junyu Ren
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Fang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Teng Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nan Wang
- Cosmos Wisdom Biotech Co. Ltd., Hangzhou, Zhejiang, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Oncology Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ming Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
LIN Z, FAN L, HE P. [Molecular Subtype of Small Cell Lung Cancer:
Challenge for Transforming into Clinical Practice]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:605-612. [PMID: 39318253 PMCID: PMC11425680 DOI: 10.3779/j.issn.1009-3419.2024.106.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 09/26/2024]
Abstract
Small cell lung cancer (SCLC), one of the histological subtypes of lung cancer, is characterized by high proliferation, early metastasis, susceptibility to drug resistance and recurrence. For several years, SCLC has always been regarded as a homogeneous disease, treated with a unified radiotherapy and chemotherapy strategy. Despite significant early therapeutic effects, drug resistance and recurrence occur quickly, and there is a lack of satisfactory treatment results, which may be due to insufficient understanding of the tumor heterogeneity of SCLC at present. Recently, the concept of SCLC molecular subtype based on the definition of relatively high expression of lineage transcription factors has been proposed in preclinical studies. This article mainly elaborates on the current status and latest findings of SCLC molecular subtype, emphasizing the potential problems that molecular typing may encounter in clinical practice, aiming to promote understanding of the research progress of molecular subtype in SCLC.
.
Collapse
|
35
|
Sen T, Takahashi N, Chakraborty S, Takebe N, Nassar AH, Karim NA, Puri S, Naqash AR. Emerging advances in defining the molecular and therapeutic landscape of small-cell lung cancer. Nat Rev Clin Oncol 2024; 21:610-627. [PMID: 38965396 PMCID: PMC11875021 DOI: 10.1038/s41571-024-00914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Small-cell lung cancer (SCLC) has traditionally been considered a recalcitrant cancer with a dismal prognosis, with only modest advances in therapeutic strategies over the past several decades. Comprehensive genomic assessments of SCLC have revealed that most of these tumours harbour deletions of the tumour-suppressor genes TP53 and RB1 but, in contrast to non-small-cell lung cancer, have failed to identify targetable alterations. The expression status of four transcription factors with key roles in SCLC pathogenesis defines distinct molecular subtypes of the disease, potentially enabling specific therapeutic approaches. Overexpression and amplification of MYC paralogues also affect the biology and therapeutic vulnerabilities of SCLC. Several other attractive targets have emerged in the past few years, including inhibitors of DNA-damage-response pathways, epigenetic modifiers, antibody-drug conjugates and chimeric antigen receptor T cells. However, the rapid development of therapeutic resistance and lack of biomarkers for effective selection of patients with SCLC are ongoing challenges. Emerging single-cell RNA sequencing data are providing insights into the plasticity and intratumoural and intertumoural heterogeneity of SCLC that might be associated with therapeutic resistance. In this Review, we provide a comprehensive overview of the latest advances in genomic and transcriptomic characterization of SCLC with a particular focus on opportunities for translation into new therapeutic approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Nobuyuki Takahashi
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Naoko Takebe
- Developmental Therapeutics Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Amin H Nassar
- Division of Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Nagla A Karim
- Inova Schar Cancer Institute Virginia, Fairfax, VA, USA
| | - Sonam Puri
- Division of Medical Oncology, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/ TSET Phase 1 program, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
36
|
Shukla V, Wang H, Varticovski L, Baek S, Wang R, Wu X, Echtenkamp F, Villa-Hernandez F, Prothro KP, Gara SK, Zhang MR, Shiffka S, Raziuddin R, Neckers LM, Linehan WM, Chen H, Hager GL, Schrump DS. Genome-Wide Analysis Identifies Nuclear Factor 1C as a Novel Transcription Factor and Potential Therapeutic Target in SCLC. J Thorac Oncol 2024; 19:1201-1217. [PMID: 38583771 DOI: 10.1016/j.jtho.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/14/2024] [Accepted: 03/30/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Recent insights regarding mechanisms mediating stemness, heterogeneity, and metastatic potential of lung cancers have yet to be fully translated to effective regimens for the treatment of these malignancies. This study sought to identify novel targets for lung cancer therapy. METHODS Transcriptomes and DNA methylomes of 14 SCLC and 10 NSCLC lines were compared with normal human small airway epithelial cells (SAECs) and induced pluripotent stem cell (iPSC) clones derived from SAEC. SCLC lines, lung iPSC (Lu-iPSC), and SAEC were further evaluated by DNase I hypersensitive site sequencing (DHS-seq). Changes in chromatin accessibility and depths of transcription factor (TF) footprints were quantified using Bivariate analysis of Genomic Footprint. Standard techniques were used to evaluate growth, tumorigenicity, and changes in transcriptomes and glucose metabolism of SCLC cells after NFIC knockdown and to evaluate NFIC expression in SCLC cells after exposure to BET inhibitors. RESULTS Considerable commonality of transcriptomes and DNA methylomes was observed between Lu-iPSC and SCLC; however, this analysis was uninformative regarding pathways unique to lung cancer. Linking results of DHS-seq to RNA sequencing enabled identification of networks not previously associated with SCLC. When combined with footprint depth, NFIC, a transcription factor not previously associated with SCLC, had the highest score of occupancy at open chromatin sites. Knockdown of NFIC impaired glucose metabolism, decreased stemness, and inhibited growth of SCLC cells in vitro and in vivo. ChIP-seq analysis identified numerous sites occupied by BRD4 in the NFIC promoter region. Knockdown of BRD4 or treatment with Bromodomain and extra-terminal domain (BET) inhibitors (BETis) markedly reduced NFIC expression in SCLC cells and SCLC PDX models. Approximately 8% of genes down-regulated by BETi treatment were repressed by NFIC knockdown in SCLC, whereas 34% of genes repressed after NFIC knockdown were also down-regulated in SCLC cells after BETi treatment. CONCLUSIONS NFIC is a key TF and possible mediator of transcriptional regulation by BET family proteins in SCLC. Our findings highlight the potential of genome-wide chromatin accessibility analysis for elucidating mechanisms of pulmonary carcinogenesis and identifying novel targets for lung cancer therapy.
Collapse
Affiliation(s)
- Vivek Shukla
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Present Address: Division of Nonclinical Sciences (DNCS), FDA, Silver Spring, Maryland
| | - Haitao Wang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lyuba Varticovski
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ruihong Wang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xinwei Wu
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank Echtenkamp
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank Villa-Hernandez
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine P Prothro
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sudheer K Gara
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary R Zhang
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie Shiffka
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Razi Raziuddin
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Leonard M Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Haobin Chen
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Present Address: Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David S Schrump
- Thoracic Epigenetics Section, Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
37
|
Peressini M, Garcia-Campelo R, Massuti B, Martí C, Cobo M, Gutiérrez V, Dómine M, Fuentes J, Majem M, de Castro J, Córdoba JF, Diz MP, Isla D, Esteban E, Carcereny E, Vila L, Moreno-Vega A, Ros S, Moreno A, García FJ, Huidobro G, Aguado C, Cebey-López V, Valdivia J, Palmero R, Lianes P, López-Brea M, Vidal OJ, Provencio M, Arriola E, Baena J, Herrera M, Bote H, Molero M, Adradas V, Ponce-Aix S, Nuñez-Buiza A, Ucero Á, Hernandez S, Lopez-Rios F, Conde E, Paz-Ares L, Zugazagoitia J. Spatially Preserved Multi-Region Transcriptomic Subtyping and Biomarkers of Chemoimmunotherapy Outcome in Extensive-Stage Small Cell Lung Cancer. Clin Cancer Res 2024; 30:3036-3049. [PMID: 38630755 DOI: 10.1158/1078-0432.ccr-24-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Transcriptomic subtyping holds promise for personalized therapy in extensive-stage small cell lung cancer (ES-SCLC). In this study, we aimed to assess intratumoral transcriptomic subtype diversity and to identify biomarkers of long-term chemoimmunotherapy benefit in human ES-SCLC. EXPERIMENTAL DESIGN We analyzed tumor samples from 58 patients with ES-SCLC enrolled in two multicenter single-arm phase IIIb studies evaluating frontline chemoimmunotherapy in Spain: n = 32 from the IMfirst trial and n = 26 from the CANTABRICO trial. We used the GeoMx Digital Spatial Profiler system to perform multi-region transcriptomic analysis. For subtype classification, we performed hierarchical clustering using the relative expression of ASCL1 (SCLC-A), NEUROD1 (SCLC-N), POU2F3 (SCLC-P), and YAP1 (SCLC-Y). RESULTS Subtype distribution was found to be similar between bothcohorts, except for SCLC-P, which was not identified in the CANTABRICO_DSP cohort. A total of 44% of the patients in both cohorts had tumors with multiple coexisting transcriptional subtypes. Transcriptional subtypes or subtype heterogeneity was not associated with outcomes. Most potential targets did not show subtype-specific expression. Consistently in both cohorts, tumors from patients with long-term benefit (time to progression ≥12 months) contained an IFNγ-dominated mRNA profile, including enhanced capacity for antigen presentation. Hypoxia and glycolytic pathways were associated with resistance to chemoimmunotherapy. CONCLUSIONS This work suggests that intratumoral heterogeneity, inconsistent association with outcome, and unclear subtype-specific target expression might be significant challenges for subtype-based precision oncology in SCLC. Preexisting IFNγ-driven immunity and mitochondrial metabolism seem to be correlates of long-term efficacy in this study, although the absence of a chemotherapy control arm precludes concluding that these are predictive features specific for immunotherapy.
Collapse
Affiliation(s)
- Melina Peressini
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | - Bartomeu Massuti
- Department of Medical Oncology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Cristina Martí
- Department of Medical Oncology, Hospital Universitario Sant Joan de Reus, Tarragona, Spain
| | - Manuel Cobo
- Department of Medical Oncology, UGC intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Vanesa Gutiérrez
- Department of Medical Oncology, Complejo Hospitalario Regional de Málaga, Málaga, Spain
| | - Manuel Dómine
- Department of Medical Oncology, Hospital Universitario Fundación Jimenez Díaz, Madrid, Spain
| | - José Fuentes
- Department of Medical Oncology, Hospital Universitario Virgen de Valme, Sevilla, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Javier de Castro
- Department of Medical Oncology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Juan F Córdoba
- Department of Medical Oncology, Hospital Universitario Arnau de Villanova, Lérida, Spain
| | - María P Diz
- Department of Medical Oncology, Complejo Asistencial Universitario de León, León, Spain
| | - Dolores Isla
- Department of Medical Oncology, Hospital Universitario Lozano Blesa, Zaragoza, Spain
| | - Emilio Esteban
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Asturias, Spain
| | - Enric Carcereny
- Department of Medical Oncology, Instituto Catalán de Oncología de Badalona, Badalona-Applied Research Group in Oncology (B-ARGO), Barcelona, Spain
| | - Laia Vila
- Department of Medical Oncology, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Moreno-Vega
- Department of Medical Oncology, Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Silverio Ros
- Department of Medical Oncology, Hospital Universitario Virgen de La Arrixaca, Murcia, Spain
| | - Amaia Moreno
- Department of Medical Oncology, Hospital de Galdakao, Vizcaya, Spain
| | - Francisco J García
- Department of Medical Oncology, Hospital Universitario Son Llatzer, Mallorca, Spain
| | - Gerardo Huidobro
- Department of Medical Oncology, Hospital Universitario Álvaro Cunqueiro, Vigo, Spain
| | - Carlos Aguado
- Department of Medical Oncology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Victor Cebey-López
- Department of Medical Oncology, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | - Javier Valdivia
- Department of Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ramón Palmero
- Department of Medical Oncology, Instituto Catalán de Oncología de Hospitalet de Llobregat, Barcelona, Spain
| | - Pilar Lianes
- Department of Medical Oncology, Hospital de Mataró, Barcelona, Spain
| | - Marta López-Brea
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Oscar J Vidal
- Department of Medical Oncology, Hospital Universitario La Fe, Valencia, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Edurne Arriola
- Department of Medical Oncology, Hospital del Mar, Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Baena
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Mercedes Herrera
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Helena Bote
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Magdalena Molero
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Vera Adradas
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Santiago Ponce-Aix
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Angel Nuñez-Buiza
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Álvaro Ucero
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
| | - Susana Hernandez
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Lopez-Rios
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Complutense University, Madrid, Spain
| | - Esther Conde
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Complutense University, Madrid, Spain
| | - Luis Paz-Ares
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| | - Jon Zugazagoitia
- Tumor Microenvironment and Immunotherapy Research Group, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Lung Cancer Clinical Research Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
38
|
Shrestha P, Kao S, Cheung VK, Cooper WA, van Zandwijk N, Rasko JEJ, Yeo D. Circulating tumor cells: advancing personalized therapy in small cell lung cancer patients. Mol Oncol 2024. [PMID: 38956984 DOI: 10.1002/1878-0261.13696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/27/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive cancer with a dismal 5-year survival of < 7%, despite the addition of immunotherapy to first-line chemotherapy. Specific tumor biomarkers, such as delta-like ligand 3 (DLL3) and schlafen11 (SLFN11), may enable the selection of more efficacious, novel immunomodulating targeted treatments like bispecific T-cell engaging monoclonal antibodies (tarlatamab) and chemotherapy with PARP inhibitors. However, obtaining a tissue biopsy sample can be challenging in SCLC. Circulating tumor cells (CTCs) have the potential to provide molecular insights into a patient's cancer through a "simple" blood test. CTCs have been studied for their prognostic ability in SCLC; however, their value in guiding treatment decisions is yet to be elucidated. This review explores novel and promising targeted therapies in SCLC, summarizes current knowledge of CTCs in SCLC, and discusses how CTCs can be utilized for precision medicine.
Collapse
Affiliation(s)
- Prajwol Shrestha
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Steven Kao
- Faculty of Medicine and Health, University of Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Veronica K Cheung
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Wendy A Cooper
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
- School of Medicine, University of Western Sydney, Australia
| | - Nico van Zandwijk
- Faculty of Medicine and Health, University of Sydney, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Concord Repatriation General Hospital, Sydney Local Health District, Concord, Australia
| | - John E J Rasko
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Dannel Yeo
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| |
Collapse
|
39
|
Ninomiya H. Resurgence of morphology: Discarded small cell lung carcinoma subtypes reflect current molecular classification. Pathol Int 2024; 74:419-421. [PMID: 38888166 DOI: 10.1111/pin.13460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Affiliation(s)
- Hironori Ninomiya
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
40
|
Scattolin D, Maso AD, Ferro A, Frega S, Bonanno L, Guarneri V, Pasello G. The emerging role of Schlafen-11 (SLFN11) in predicting response to anticancer treatments: Focus on small cell lung cancer. Cancer Treat Rev 2024; 128:102768. [PMID: 38797062 DOI: 10.1016/j.ctrv.2024.102768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Small cell lung cancer (SCLC) is characterized by a dismal prognosis. Many efforts have been made so far for identifying novel biomarkers for a personalized treatment for SCLC patients. Schlafen 11 (SLFN11) is a protein differently expressed in many cancers and recently emerged as a new potential biomarker. Lower expression of SLFN11 correlates with a worse prognosis in SCLC and other tumors. SLFN11 has a role in tumorigenesis, inducing replication arrest in the presence of DNA damage through the block of the replication fork. SLFN11 interacts also with chromatin accessibility, proteotoxic stress and mammalian target of rapamycin signalling pathway. The expression of SLFN11 is regulated by epigenetic mechanisms, including promoter methylation, histone deacetylation, and the histone methylation. The downregulation of SLFN11 correlates with a worse response to topoisomerase I and II inhibitors, alkylating agents, and poly ADP-ribose polymerase inhibitors in different cancer types. Some studies exploring strategies for overcoming drug resistance in tumors with low levels of SLFN11 showed promising results. One of these strategies includes the interaction with the Ataxia Telangiectasia and Rad3-related pathway, constitutively activated and leading to cell survival and tumor growth in the presence of low levels of SLFN11. Furthermore, the expression of SLFN11 is dynamic through time and different anticancer therapy and liquid biopsy seems to be an attractive tool for catching SLFN11 different expressions. Despite this, further investigations exploring SLFN11 as a predictive biomarker, its longitudinal changes, and new strategies to overcome drug resistances are needed.
Collapse
Affiliation(s)
- Daniela Scattolin
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Alessandra Ferro
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Stefano Frega
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Laura Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.
| |
Collapse
|
41
|
Desai P, Takahashi N, Kumar R, Nichols S, Malin J, Hunt A, Schultz C, Cao Y, Tillo D, Nousome D, Chauhan L, Sciuto L, Jordan K, Rajapakse V, Tandon M, Lissa D, Zhang Y, Kumar S, Pongor L, Singh A, Schroder B, Sharma AK, Chang T, Vilimas R, Pinkiert D, Graham C, Butcher D, Warner A, Sebastian R, Mahon M, Baker K, Cheng J, Berger A, Lake R, Abel M, Krishnamurthy M, Chrisafis G, Fitzgerald P, Nirula M, Goyal S, Atkinson D, Bateman NW, Abulez T, Nair G, Apolo A, Guha U, Karim B, El Meskini R, Ohler ZW, Jolly MK, Schaffer A, Ruppin E, Kleiner D, Miettinen M, Brown GT, Hewitt S, Conrads T, Thomas A. Microenvironment shapes small-cell lung cancer neuroendocrine states and presents therapeutic opportunities. Cell Rep Med 2024; 5:101610. [PMID: 38897168 PMCID: PMC11228806 DOI: 10.1016/j.xcrm.2024.101610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/04/2023] [Accepted: 05/17/2024] [Indexed: 06/21/2024]
Abstract
Small-cell lung cancer (SCLC) is the most fatal form of lung cancer. Intratumoral heterogeneity, marked by neuroendocrine (NE) and non-neuroendocrine (non-NE) cell states, defines SCLC, but the cell-extrinsic drivers of SCLC plasticity are poorly understood. To map the landscape of SCLC tumor microenvironment (TME), we apply spatially resolved transcriptomics and quantitative mass spectrometry-based proteomics to metastatic SCLC tumors obtained via rapid autopsy. The phenotype and overall composition of non-malignant cells in the TME exhibit substantial variability, closely mirroring the tumor phenotype, suggesting TME-driven reprogramming of NE cell states. We identify cancer-associated fibroblasts (CAFs) as a crucial element of SCLC TME heterogeneity, contributing to immune exclusion, and predicting exceptionally poor prognosis. Our work provides a comprehensive map of SCLC tumor and TME ecosystems, emphasizing their pivotal role in SCLC's adaptable nature, opening possibilities for reprogramming the TME-tumor communications that shape SCLC tumor states.
Collapse
Affiliation(s)
- Parth Desai
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Medical Oncology, Fox Chase Cancer Center, Temple University Hospital and Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Nobuyuki Takahashi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Rajesh Kumar
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samantha Nichols
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin Malin
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Allison Hunt
- Women's Health Integrated Research Center, Inova Health System, Falls Church, VA, USA
| | - Christopher Schultz
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yingying Cao
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Desiree Tillo
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Darryl Nousome
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lakshya Chauhan
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Linda Sciuto
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly Jordan
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Vinodh Rajapakse
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mayank Tandon
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Delphine Lissa
- Laboratory of Human Carcinogenesis, Center for Cancer Research National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yang Zhang
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suresh Kumar
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lorinc Pongor
- HCEMM Cancer Genomics and Epigenetics Research Group, Szeged, Hungary
| | - Abhay Singh
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Brett Schroder
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ajit Kumar Sharma
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiangen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rasa Vilimas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Pinkiert
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chante Graham
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Andrew Warner
- Molecular Histopathology Laboratory, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Robin Sebastian
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mimi Mahon
- Pain and Palliative care services, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Karen Baker
- Pain and Palliative care services, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Jennifer Cheng
- Pain and Palliative care services, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Ann Berger
- Pain and Palliative care services, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Ross Lake
- Laboratory of Genitourinary cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Melissa Abel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Manan Krishnamurthy
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - George Chrisafis
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter Fitzgerald
- CCR Collaborative Bioinformatics, Resource, Office of Science and Technology Resources, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Micheal Nirula
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shubhank Goyal
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Devon Atkinson
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Nicholas W Bateman
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD, USA
| | - Tamara Abulez
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD, USA
| | - Govind Nair
- National Institute of Neurological Disorders and Stroke, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Udayan Guha
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Rajaa El Meskini
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Zoe Weaver Ohler
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Mohit Kumar Jolly
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Alejandro Schaffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David Kleiner
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - G Tom Brown
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen Hewitt
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Conrads
- Women's Health Integrated Research Center, Inova Health System, Falls Church, VA, USA
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
Jimenez L, Stolzenbach V, Ozawa PMM, Ramirez-Solano M, Liu Q, Sage J, Weaver AM. Extracellular vesicles from non-neuroendocrine SCLC cells promote adhesion and survival of neuroendocrine SCLC cells. Proteomics 2024; 24:e2300030. [PMID: 37926756 PMCID: PMC11648350 DOI: 10.1002/pmic.202300030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
Small cell lung cancer (SCLC) tumors are made up of distinct cell subpopulations, including neuroendocrine (NE) and non-neuroendocrine (non-NE) cells. While secreted factors from non-NE SCLC cells have been shown to support the growth of the NE cells, the underlying molecular factors are not well understood. Here, we show that exosome-type small extracellular vesicles (SEVs) secreted from non-NE SCLC cells promote adhesion and survival of NE SCLC cells. Proteomic analysis of purified SEVs revealed that extracellular matrix (ECM) proteins and integrins are highly enriched in SEVs of non-NE cells whereas nucleic acid-binding proteins are enriched in SEVs purified from NE cells. Addition of select purified ECM proteins identified in purified extracellular vesicles (EVs), specifically fibronectin, laminin 411, and laminin 511, were able to substitute for the role of non-NE-derived SEVs in promoting adhesion and survival of NE SCLC cells. Those same proteins were differentially expressed by human SCLC subtypes. These data suggest that ECM-carrying SEVs secreted by non-NE cells play a key role in supporting the growth and survival of NE SCLC cells.
Collapse
Affiliation(s)
- Lizandra Jimenez
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Center for Extracellular Vesicle Research, Vanderbilt University, Nashville, Tennessee
| | - Victor Stolzenbach
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Center for Extracellular Vesicle Research, Vanderbilt University, Nashville, Tennessee
| | - Patricia M. M. Ozawa
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Center for Extracellular Vesicle Research, Vanderbilt University, Nashville, Tennessee
| | | | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julien Sage
- Department of Pediatrics, Stanford Medicine, Stanford, California
- Department of Genetics, Stanford Medicine, Stanford, California
| | - Alissa M. Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Center for Extracellular Vesicle Research, Vanderbilt University, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
43
|
Hayashi T. Genetic diversity in small cell lung carcinoma. Transl Lung Cancer Res 2024; 13:1169-1172. [PMID: 38854933 PMCID: PMC11157378 DOI: 10.21037/tlcr-24-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/22/2024] [Indexed: 06/11/2024]
Affiliation(s)
- Takuo Hayashi
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Diagnostic Pathology Center, Juntendo Hospital, Bunkyo-ku, Tokyo, Japan
- Bioresource Research Center, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
44
|
Xie M, Vuko M, Rodriguez-Canales J, Zimmermann J, Schick M, O'Brien C, Paz-Ares L, Goldman JW, Garassino MC, Gay CM, Heymach JV, Jiang H, Barrett JC, Stewart RA, Lai Z, Byers LA, Rudin CM, Shrestha Y. Molecular classification and biomarkers of outcome with immunotherapy in extensive-stage small-cell lung cancer: analyses of the CASPIAN phase 3 study. Mol Cancer 2024; 23:115. [PMID: 38811992 PMCID: PMC11137956 DOI: 10.1186/s12943-024-02014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND We explored potential predictive biomarkers of immunotherapy response in patients with extensive-stage small-cell lung cancer (ES-SCLC) treated with durvalumab (D) + tremelimumab (T) + etoposide-platinum (EP), D + EP, or EP in the randomized phase 3 CASPIAN trial. METHODS 805 treatment-naïve patients with ES-SCLC were randomized (1:1:1) to receive D + T + EP, D + EP, or EP. The primary endpoint was overall survival (OS). Patients were required to provide an archived tumor tissue block (or ≥ 15 newly cut unstained slides) at screening, if these samples existed. After assessment for programmed cell death ligand-1 expression and tissue tumor mutational burden, residual tissue was used for additional molecular profiling including by RNA sequencing and immunohistochemistry. RESULTS In 182 patients with transcriptional molecular subtyping, OS with D ± T + EP was numerically highest in the SCLC-inflamed subtype (n = 10, median 24.0 months). Patients derived benefit from immunotherapy across subtypes; thus, additional biomarkers were investigated. OS benefit with D ± T + EP versus EP was greater with high versus low CD8A expression/CD8 cell density by immunohistochemistry, but with no additional benefit with D + T + EP versus D + EP. OS benefit with D + T + EP versus D + EP was associated with high expression of CD4 (median 25.9 vs. 11.4 months) and antigen-presenting and processing machinery (25.9 vs. 14.6 months) and MHC I and II (23.6 vs. 17.3 months) gene signatures, and with higher MHC I expression by immunohistochemistry. CONCLUSIONS These findings demonstrate the tumor microenvironment is important in mediating better outcomes with D ± T + EP in ES-SCLC, with canonical immune markers associated with hypothesized immunotherapy mechanisms of action defining patient subsets that respond to D ± T. TRIAL REGISTRATION ClinicalTrials.gov, NCT03043872.
Collapse
Affiliation(s)
- Mingchao Xie
- Oncology Data Science, AstraZeneca, Waltham, MA, USA
| | - Miljenka Vuko
- Computational Pathology, AstraZeneca, Munich, Germany
| | | | | | - Markus Schick
- Computational Pathology, AstraZeneca, Munich, Germany
| | - Cathy O'Brien
- Biostatistics, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Marina Chiara Garassino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- The University of Chicago, Chicago, IL, USA
| | - Carl M Gay
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John V Heymach
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haiyi Jiang
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - J Carl Barrett
- Translational Medicine, AstraZeneca, Waltham, MA, United States
| | - Ross A Stewart
- Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Zhongwu Lai
- Oncology Data Science, AstraZeneca, Waltham, MA, USA
| | - Lauren A Byers
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Yashaswi Shrestha
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
45
|
Trillo Aliaga P, Del Signore E, Fuorivia V, Spitaleri G, Asnaghi R, Attili I, Corvaja C, Carnevale Schianca A, Passaro A, de Marinis F. The Evolving Scenario of ES-SCLC Management: From Biology to New Cancer Therapeutics. Genes (Basel) 2024; 15:701. [PMID: 38927637 PMCID: PMC11203015 DOI: 10.3390/genes15060701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine carcinoma accounting for 15% of lung cancers with dismal survival outcomes. Minimal changes in therapy and prognosis have occurred in SCLC for the past four decades. Recent progress in the treatment of extensive-stage disease (ES-SCLC) has been marked by incorporating immune checkpoint inhibitors (ICIs) into platinum-based chemotherapy, leading to modest improvements. Moreover, few second-line-and-beyond treatment options are currently available. The main limitation for the molecular study of SCLC has been the scarcity of samples, because only very early diseases are treated with surgery and biopsies are not performed when the disease progresses. Despite all these difficulties, in recent years we have come to understand that SCLC is not a homogeneous disease. At the molecular level, in addition to the universal loss of retinoblastoma (RB) and TP53 genes, a recent large molecular study has identified other mutations that could serve as targets for therapy development or patient selection. In recent years, there has also been the identification of new genetic subtypes which have shown us how intertumor heterogeneity exists. Moreover, SCLC can also develop intratumoral heterogeneity linked mainly to the concept of cellular plasticity, mostly due to the development of resistance to therapies. The aim of this review is to quickly present the current standard of care of ES-SCLC, to focus on the molecular landscapes and subtypes of SCLC, subsequently present the most promising therapeutic strategies under investigation, and finally recap the future directions of ongoing clinical trials for this aggressive disease which still remains a challenge.
Collapse
Affiliation(s)
- Pamela Trillo Aliaga
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Ester Del Signore
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Valeria Fuorivia
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Riccardo Asnaghi
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Ilaria Attili
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Carla Corvaja
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Ambra Carnevale Schianca
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Haematology (DIPO), University of Milan, 20122 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
46
|
Ito T. Molecular pathology of small cell lung cancer: Overview from studies on neuroendocrine differentiation regulated by ASCL1 and Notch signaling. Pathol Int 2024; 74:239-251. [PMID: 38607250 DOI: 10.1111/pin.13426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
Pulmonary neuroendocrine (NE) cells are rare airway epithelial cells. The balance between Achaete-scute complex homolog 1 (ASCL1) and hairy and enhancer of split 1, one of the target molecules of the Notch signaling pathway, is crucial for NE differentiation. Small cell lung cancer (SCLC) is a highly aggressive lung tumor, characterized by rapid cell proliferation, a high metastatic potential, and the acquisition of resistance to treatment. The subtypes of SCLC are defined by the expression status of NE cell-lineage transcription factors, such as ASCL1, which roles are supported by SRY-box 2, insulinoma-associated protein 1, NK2 homeobox 1, and wingless-related integration site signaling. This network reinforces NE differentiation and may induce the characteristic morphology and chemosensitivity of SCLC. Notch signaling mediates cell-fate decisions, resulting in an NE to non-NE fate switch. The suppression of NE differentiation may change the histological type of SCLC to a non-SCLC morphology. In SCLC with NE differentiation, Notch signaling is typically inactive and genetically or epigenetically regulated. However, Notch signaling may be activated after chemotherapy, and, in concert with Yes-associated protein signaling and RE1-silencing transcription factor, suppresses NE differentiation, producing intratumor heterogeneity and chemoresistance. Accumulated information on the molecular mechanisms of SCLC will contribute to further advances in the control of this recalcitrant cancer.
Collapse
Grants
- 20H03691 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 18K19489 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 16590318 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 25460439 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- Smoking Research Foundation, Japan
Collapse
Affiliation(s)
- Takaaki Ito
- Department of Medical Technology, Kumamoto Health Science University Faculty of Health Sciences, Kumamoto, Japan
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
47
|
Ng J, Cai L, Girard L, Prall OW, Rajan N, Khoo C, Batrouney A, Byrne DJ, Boyd DK, Kersbergen AJ, Christie M, Minna JD, Burr ML, Sutherland KD. Molecular and Pathologic Characterization of YAP1-Expressing Small Cell Lung Cancer Cell Lines Leads to Reclassification as SMARCA4-Deficient Malignancies. Clin Cancer Res 2024; 30:1846-1858. [PMID: 38180245 PMCID: PMC11061608 DOI: 10.1158/1078-0432.ccr-23-2360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/08/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
PURPOSE The classification of small cell lung cancer (SCLC) into distinct molecular subtypes defined by ASCL1, NEUROD1, POU2F3, or YAP1 (SCLC-A, -N, -P, or -Y) expression, paves the way for a personalized treatment approach. However, the existence of a distinct YAP1-expressing SCLC subtype remains controversial. EXPERIMENTAL DESIGN To better understand YAP1-expressing SCLC, the mutational landscape of human SCLC cell lines was interrogated to identify pathogenic alterations unique to SCLC-Y. Xenograft tumors, generated from cell lines representing the four SCLC molecular subtypes, were evaluated by a panel of pathologists who routinely diagnose thoracic malignancies. Diagnoses were complemented by transcriptomic analysis of primary tumors and human cell line datasets. Protein expression profiles were validated in patient tumor tissue. RESULTS Unexpectedly, pathogenic mutations in SMARCA4 were identified in six of eight SCLC-Y cell lines and correlated with reduced SMARCA4 mRNA and protein expression. Pathologist evaluations revealed that SMARCA4-deficient SCLC-Y tumors exhibited features consistent with thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT). Similarly, the transcriptional profile SMARCA4-mutant SCLC-Y lines more closely resembled primary SMARCA4-UT, or SMARCA4-deficient non-small cell carcinoma, than SCLC. Furthermore, SMARCA4-UT patient samples were associated with a YAP1 transcriptional signature and exhibited strong YAP1 protein expression. Together, we found little evidence to support a diagnosis of SCLC for any of the YAP1-expressing cell lines originally used to define the SCLC-Y subtype. CONCLUSIONS SMARCA4-mutant SCLC-Y cell lines exhibit characteristics consistent with SMARCA4-deficient malignancies rather than SCLC. Our findings suggest that, unlike ASCL1, NEUROD1, and POU2F3, YAP1 is not a subtype defining transcription factor in SCLC. See related commentary by Rekhtman, p. 1708.
Collapse
Affiliation(s)
- Jin Ng
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ling Cai
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, Texas
- Children's Research Institute, UT Southwestern Medical Center, Dallas, Texas
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Luc Girard
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Owen W.J. Prall
- Department of Anatomical Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Neeha Rajan
- Department of Anatomical Pathology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Christine Khoo
- Department of Anatomical Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ahida Batrouney
- Department of Anatomical Pathology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - David J. Byrne
- Department of Anatomical Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Danielle K. Boyd
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Ariena J. Kersbergen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Michael Christie
- Department of Anatomical Pathology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - John D. Minna
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | - Marian L. Burr
- Division of Genome Science and Cancer, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- Department of Anatomical Pathology, ACT Pathology, Canberra Health Services, Canberra, Australian Capital Territory, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Kate D. Sutherland
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
48
|
Fűr GM, Nemes K, Magó É, Benő AÁ, Topolcsányi P, Moldvay J, Pongor LS. Applied models and molecular characteristics of small cell lung cancer. Pathol Oncol Res 2024; 30:1611743. [PMID: 38711976 PMCID: PMC11070512 DOI: 10.3389/pore.2024.1611743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024]
Abstract
Small cell lung cancer (SCLC) is a highly aggressive type of cancer frequently diagnosed with metastatic spread, rendering it surgically unresectable for the majority of patients. Although initial responses to platinum-based therapies are often observed, SCLC invariably relapses within months, frequently developing drug-resistance ultimately contributing to short overall survival rates. Recently, SCLC research aimed to elucidate the dynamic changes in the genetic and epigenetic landscape. These have revealed distinct subtypes of SCLC, each characterized by unique molecular signatures. The recent understanding of the molecular heterogeneity of SCLC has opened up potential avenues for precision medicine, enabling the development of targeted therapeutic strategies. In this review, we delve into the applied models and computational approaches that have been instrumental in the identification of promising drug candidates. We also explore the emerging molecular diagnostic tools that hold the potential to transform clinical practice and patient care.
Collapse
Affiliation(s)
- Gabriella Mihalekné Fűr
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Kolos Nemes
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Éva Magó
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
- Genome Integrity and DNA Repair Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Alexandra Á. Benő
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Petronella Topolcsányi
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Judit Moldvay
- Department of Pulmonology, Szeged University Szent-Gyorgyi Albert Medical School, Szeged, Hungary
- 1st Department of Pulmonology, National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Lőrinc S. Pongor
- Cancer Genomics and Epigenetics Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| |
Collapse
|
49
|
Yao Z, Song P, Jiao W. Pathogenic role of super-enhancers as potential therapeutic targets in lung cancer. Front Pharmacol 2024; 15:1383580. [PMID: 38681203 PMCID: PMC11047458 DOI: 10.3389/fphar.2024.1383580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Lung cancer is still one of the deadliest malignancies today, and most patients with advanced lung cancer pass away from disease progression that is uncontrollable by medications. Super-enhancers (SEs) are large clusters of enhancers in the genome's non-coding sequences that actively trigger transcription. Although SEs have just been identified over the past 10 years, their intricate structure and crucial role in determining cell identity and promoting tumorigenesis and progression are increasingly coming to light. Here, we review the structural composition of SEs, the auto-regulatory circuits, the control mechanisms of downstream genes and pathways, and the characterization of subgroups classified according to SEs in lung cancer. Additionally, we discuss the therapeutic targets, several small-molecule inhibitors, and available treatment options for SEs in lung cancer. Combination therapies have demonstrated considerable advantages in preclinical models, and we anticipate that these drugs will soon enter clinical studies and benefit patients.
Collapse
Affiliation(s)
- Zhiyuan Yao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Song
- Department of Thoracic Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
50
|
Hiatt JB, Doebley AL, Arnold HU, Adil M, Sandborg H, Persse TW, Ko M, Wu F, Quintanal Villalonga A, Santana-Davila R, Eaton K, Dive C, Rudin CM, Thomas A, Houghton AM, Ha G, MacPherson D. Molecular phenotyping of small cell lung cancer using targeted cfDNA profiling of transcriptional regulatory regions. SCIENCE ADVANCES 2024; 10:eadk2082. [PMID: 38598634 PMCID: PMC11006233 DOI: 10.1126/sciadv.adk2082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/27/2024] [Indexed: 04/12/2024]
Abstract
We report an approach for cancer phenotyping based on targeted sequencing of cell-free DNA (cfDNA) for small cell lung cancer (SCLC). In SCLC, differential activation of transcription factors (TFs), such as ASCL1, NEUROD1, POU2F3, and REST defines molecular subtypes. We designed a targeted capture panel that identifies chromatin organization signatures at 1535 TF binding sites and 13,240 gene transcription start sites and detects exonic mutations in 842 genes. Sequencing of cfDNA from SCLC patient-derived xenograft models captured TF activity and gene expression and revealed individual highly informative loci. Prediction models of ASCL1 and NEUROD1 activity using informative loci achieved areas under the receiver operating characteristic curve (AUCs) from 0.84 to 0.88 in patients with SCLC. As non-SCLC (NSCLC) often transforms to SCLC following targeted therapy, we applied our framework to distinguish NSCLC from SCLC and achieved an AUC of 0.99. Our approach shows promising utility for SCLC subtyping and transformation monitoring, with potential applicability to diverse tumor types.
Collapse
Affiliation(s)
- Joseph B. Hiatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Veterans Affairs Puget Sound Healthcare System - Seattle Branch, Seattle, WA, USA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Anna-Lisa Doebley
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Henry U. Arnold
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mohamed Adil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Holly Sandborg
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas W. Persse
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Minjeong Ko
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alvaro Quintanal Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rafael Santana-Davila
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Keith Eaton
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Caroline Dive
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Charles M. Rudin
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Graduate Program in Pharmacology, Weill Cornell Medical College; New York, NY, USA
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - A. McGarry Houghton
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gavin Ha
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|