1
|
Sajic T, Vizovišek M, Arni S, Ciuffa R, Mehnert M, Lenglet S, Weder W, Gallart-Ayala H, Ivanisevic J, Buljan M, Thomas A, Hillinger S, Aebersold R. Depletion-dependent activity-based protein profiling using SWATH/DIA-MS detects serine hydrolase lipid remodeling in lung adenocarcinoma progression. Nat Commun 2025; 16:4889. [PMID: 40425563 PMCID: PMC12117057 DOI: 10.1038/s41467-025-59564-x] [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: 11/16/2023] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Systematic inference of enzyme activity in human tumors is key to understanding cancer progression and resistance to therapy. However, standard protein or transcript abundances are blind to the activity status of the measured enzymes, regulated, for example, by active-site amino acid mutations or post-translational protein modifications. Current methods for activity-based proteome profiling (ABPP), which combine mass spectrometry (MS) with chemical probes, quantify the fraction of enzymes that are catalytically active. Here, we describe depletion-dependent ABPP (dd-ABPP) combined with automated SWATH/DIA-MS, which simultaneously determines three molecular layers of studied enzymes: i) catalytically active enzyme fractions, ii) enzyme and background protein abundances, and iii) context-dependent enzyme-protein interactions. We demonstrate the utility of the method in advanced lung adenocarcinoma (LUAD) by monitoring nearly 4000 protein groups and 200 serine hydrolases (SHs) in tumor and adjacent tissue sections routinely collected for patient histopathology. The activity profiles of 23 SHs and the abundance of 59 proteins associated with these enzymes retrospectively classified aggressive LUAD. The molecular signature revealed accelerated lipoprotein depalmitoylation via palmitoyl(protein)hydrolase activities, further confirmed by excess palmitate and its metabolites. The approach is universal and applicable to other enzyme families with available chemical probes, providing clinicians with a biochemical rationale for tumor sample classification.
Collapse
Affiliation(s)
- Tatjana Sajic
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | - Matej Vizovišek
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Stephan Arni
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland
| | - Rodolfo Ciuffa
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Martin Mehnert
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland
| | - Sébastien Lenglet
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics and Lipidomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV, Rue du Bugnon 19, CH-1005, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics and Lipidomics Platform, Faculty of Biology and Medicine, University of Lausanne, Quartier UNIL-CHUV, Rue du Bugnon 19, CH-1005, Lausanne, Switzerland
| | - Marija Buljan
- Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St Gallen, Dübendorf, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Aurelien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Sven Hillinger
- Division of Thoracic Surgery, University Hospital Zurich (UHZ), Zürich, Switzerland.
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH, Zurich, Switzerland.
- Faculty of Science, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Dong Y, Wang X, Dong C, Li P, Liu Z, Tian X. Characteristics of folic acid metabolism-related genes unveil prognosis and treatment strategy in lung adenocarcinoma. BMC Pulm Med 2025; 25:255. [PMID: 40405133 PMCID: PMC12101037 DOI: 10.1186/s12890-025-03694-x] [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: 04/27/2024] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer. Folic acid metabolism-related genes (FAMGs) have received increased attention because of their distinct role in DNA synthesis and repair. Nevertheless, the function of FAMGs in LUAD remains ambiguous. METHODS LUAD transcriptome data from GEO and TCGA were analyzed. Patients were classified into two clusters based on gene expression levels, revealing distinct overall survival (OS) outcomes. Common differentially expressed genes (DEGs) were identified between LUAD and normal tissues, as well as between the two clusters. A prognostic risk model was established using Cox regression analysis to predict outcomes of LUAD patients and was validated with Kaplan-Meier and ROC curve analysis. Clinical correlations and enrichment analyses were carried out to explore the functions of DEGs and their associations with clinical characteristics of LUAD patients. The tumor microenvironment and drug sensitivity were evaluated between two risk subgroups. Moreover, expression levels of prognostic genes were validated across datasets using the Wilcoxon-test. RESULTS The study identified seventy-seven common DEGs and nine prognostic genes (ANLN, PLK1, DLGAP5, PRC1, CYP4B1, MKI67, KIF23, BIRC5, TK1). The risk model could effectively predict the prognosis of LUAD patients. Clinical correlation analysis revealed that age, pathologic-T, pathologic-N, and tumor stage were significantly correlated with the risk score. Enrichment analysis showed that DEGs between the two risk subgroups were predominantly enriched in cell cycle and cellular senescence pathways. Differences in immune cell infiltration and immunotherapy markers were markedly noted between the two risk subgroups. Drug sensitivity analysis disclosed significantly diverse responses to sixty-eight drugs between the two risk subgroups. Consistent expression tendencies of prognostic genes were observed across datasets. CONCLUSION The prognostic model based on FAMGs demonstrates considerable potential for guiding diagnosis and clinical management of LUAD patients.
Collapse
Affiliation(s)
- Yanting Dong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoyan Wang
- Beijing Health Vocational College, Beijing, China
| | - Chuanchuan Dong
- Clinical Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Peiqi Li
- Clinical Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhuola Liu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinrui Tian
- Department of Geratology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
El Gazzah E, Parker S, Pierobon M. Multi-omic profiling in breast cancer: utility for advancing diagnostics and clinical care. Expert Rev Mol Diagn 2025; 25:165-181. [PMID: 40193192 DOI: 10.1080/14737159.2025.2482639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Breast cancer remains a major global health challenge. While advances in precision oncology have contributed to improvements in patient outcomes and provided a deeper understanding of the biological mechanisms that drive the disease, historically, research and patients' allocation to treatment have heavily relied on single-omic approaches, analyzing individual molecular dimensions such as genomics, transcriptomics, or proteomics. While these have provided deep insights into breast cancer biology, they often fail to offer a complete understanding of the disease's complex molecular landscape. AREAS COVERED In this review, the authors explore the recent advancements in multi-omic research in the realm of breast cancer and use clinical data to show how multi-omic integration can offer a more holistic understanding of the molecular alterations and their functional consequences underlying breast cancer. EXPERT OPINION The overall developments in multi-omic research and AI are expected to complement precision diagnostics through potentially refining prognostic models, and treatment selection. Overcoming challenges such as cost, data complexity, and lack of standardization is crucial for unlocking the full potential of multi-omics and AI in breast cancer patient care to enable the advancement of personalized treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Emna El Gazzah
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Scott Parker
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Mariaelena Pierobon
- School of Systems Biology, George Mason University, Manassas, VA, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| |
Collapse
|
4
|
Lin YD, Li HJ, Hong HZ, Qi YF, Li YY, Yang XN, Wu YL, Zhong WZ. Genomic profiling of aggressive pathologic features in lung adenocarcinoma. Lung Cancer 2025; 203:108460. [PMID: 40179539 DOI: 10.1016/j.lungcan.2025.108460] [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: 05/30/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Pathologic features involving LVI (lympho-vascular invasion), PNI (perineural invasion), STAS (spread through air spaces), and Grade 3 pattern (from the International Association for the Study of Lung Cancer grading system) are related to having an aggressive phenotype and linked to poor prognosis. However, few studies have conducted in-depth analyses of these features simultaneously with genomic profiling. METHODS A total of 1559 sequencing of adenocarcinoma samples were included in the common driver mutations analysis, 1306 samples were brought into genomic mapping analysis. OncoSG's East Asian ancestry dataset was implemented for Tumor-Node-Metastasis-Biomarker (TNMB) classification and prognostic assessment. RESULTS EGFR was more significantly prevalent in LVI negativity (P = 0.021), STAS negativity (P = 0.002), and moderate grade (P < 0.001). ALK was significantly interrelated with LVI (P = 0.028), STAS (P < 0.001), and poor grade (P < 0.001); ROS1 and STAS positivity (P = 0.031), poor grade (P = 0.016) were significantly related. KRAS (P = 0.003) and BRAF-V600E (P = 0.002) were only significantly intertwined with poor grade. Apart from common driver mutations, TP53, CHEK2, KEAP1, PTEN, RB1, NF1 were significantly enriched in LVI samples (P < 0.05). TP53, PTEN, CTNNB1, HGF, NF1 were more prominent in STAS (P < 0.01). TP53, LRP1B, NF1 were significantly more prevalent in Grade 3 pattern (P < 0.001). The mixture of STK11, PTEN, and TOP2A generated by exclusive mutations may be a potential predictor of TNMB categorization towards survival. The HR of stage II compared I of TNMB was 2.28 (95 % CI 1.36-3.86, P < 0.001), while stage III compared II was 1.95 (95 % CI 1.04-3.21, P = 0.031). CONCLUSIONS This analysis demonstrated the correlation of pathologic features with common driver mutations, key mutations and canonical oncogenic signaling pathways. The data highlighted the similarities and differences among these features horizontally, and provide new insights in TNMB classification and prognostic assessment.
Collapse
Affiliation(s)
- Yi-Duo Lin
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hong-Ji Li
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hui-Zhao Hong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Fan Qi
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yun-Yi Li
- School of Software Engineering, South China University of Technology, Guangzhou, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Lin H, Hua J, Gong Z, Chen M, Qiu B, Wu Y, He W, Wang Y, Feng Z, Liang Y, Long W, Li R, Kuang Q, Chen Y, Lu J, Luo S, Zhao W, Yan L, Chen X, Shi Z, Xu Z, Mo Z, Liu E, Han C, Cui Y, Yang X, Chen X, Liu J, Pan X, Madabhushi A, Lu C, Liu Z. Multimodal radiopathological integration for prognosis and prediction of adjuvant chemotherapy benefit in resectable lung adenocarcinoma: A multicentre study. Cancer Lett 2025; 616:217557. [PMID: 39954935 DOI: 10.1016/j.canlet.2025.217557] [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: 11/30/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Lung adenocarcinoma (LUAD) has a heterogeneous prognosis and controversial postoperative treatment protocols. We aim to develop and validate a multimodal analysis framework that integrates CT images with H&E-stained whole-slide images (WSIs) to enhance risk stratification and predict adjuvant chemotherapy benefit in LUAD patients. We retrospectively collected data from 1039 resectable LUAD patients (stage I-III) across four centres, forming a training dataset (n = 303), two testing datasets (n = 197 and n = 228) for survival analysis, and a feature testing dataset (n = 311) for interpretability analysis. We extracted 487 tumour/peritumour radiomics features from CT images and 783 multiscale pathomics features from WSIs, characterising the shape of tumour (CT) and cancer nuclei (WSIs), as well as the intensity and texture of tumour/peritumour regions (CT) and tumour regions/epithelium/stroma (WSIs). A survival support vector machine (SVM) was employed to establish a radiopathomics signature using the optimal set of multimodal features, including 2 tumour radiomics features, 3 peritumour radiomics features, and 4 nuclei heterogeneity pathomics features. The radiopathomics signature outperformed both radiomics and pathomics signatures in predicting disease-free survival (DFS) (C-index: training dataset, 0.744 vs. 0.734 and 0.692; testing dataset 1, 0.719 vs. 0.701 and 0.638; testing dataset 2, 0.711 vs. 0.689 and 0.684), demonstrating greater robustness compared to the state-of-the-art deep learning integration approaches. It provided additional prognostic information beyond clinical risk factors (C-index of clinical plus radiopathomics vs. clinical models: training dataset, 0.763 vs. 0.676; testing dataset 1, 0.739 vs. 0.676; testing dataset 2, 0.711 vs. 0.699, p < 0.001). Compared to low-risk patients categorised by the radiopathomics signature, high-risk patients achieved comparable DFS when receiving adjuvant chemotherapy (training dataset, HR = 1.53, 95 % CI 0.85-2.73, p = 0.153; testing dataset 1 and 2, HR = 1.62, 95 % CI 0.92-2.85, p = 0.096), but had significantly worse DFS when only observed after surgery (training dataset, HR = 4.46, 95 % CI 2.82-7.05, p < 0.001; testing datasets 1 and 2, HR = 3.52, 95 % CI 2.26-5.49, p < 0.001), indicating the predictive value of the radiopathomics signature for adjuvant chemotherapy benefit (interaction p < 0.05). Further interpretability analysis revealed that the radiopathomics signature was associated with various prognostic/treatment-related biomarkers, including differentiation, immune phenotypes, and EGFR status. The multimodal integration framework offered a cost-effective approach for LUAD characterisation by leveraging complementary information from radiological and histopathological imaging. The radiopathomics signature demonstrated robust prognostic capabilities, providing valuable insights for postoperative treatment decisions.
Collapse
Affiliation(s)
- Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Junjie Hua
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhengze Gong
- Information and Data Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Mingwei Chen
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Bingjiang Qiu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, 510080, China
| | - Yuxin Wu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Wenfeng He
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Yumeng Wang
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Zhengyun Feng
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wansheng Long
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Ronggang Li
- Department of Pathology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Qionglian Kuang
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Yingxin Chen
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jiawei Lu
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Shiwei Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Zhenwei Shi
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zeyan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Ziyang Mo
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Entao Liu
- WeiLun PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chu Han
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yanfen Cui
- Department of Radiology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xiaotang Yang
- Department of Radiology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China.
| | - Xiangmeng Chen
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China.
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Anant Madabhushi
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| | - Cheng Lu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Zhou DDX, Dalrymple J, Klingberg D, Lin FPY, Lord SJ, Cooper WA, Zaheed M, Simes RJ, John T, Lee CK. Clinical Impact of Somatic Genomic Variants of Oncogenes and Tumor Suppressor Genes in Previously Treated Advanced Non-Small Cell Lung Cancer. JCO Precis Oncol 2025; 9:e2400673. [PMID: 40239138 DOI: 10.1200/po-24-00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Next-generation sequencing in non-small cell lung cancer (NSCLC) identifies somatic genomic variants (SGVs) in cancer susceptibility genes (CSGs). We hypothesized that SGVs would be associated with poorer overall survival (OS) but greater benefit with immune checkpoint inhibitors over chemotherapy. We investigated the prevalence and predictive value of SGVs, using data from OAK and POPLAR trials comparing atezolizumab with docetaxel. METHODS We curated a list of SGVs (excluding TP53, EGFR, ALK, and ROS1) on the basis of CSGs associated with tumorigenesis. We classified participants as SGV mutant or wild-type using baseline plasma analyzed by the FoundationOne Liquid CDx assay. Cox regression analyses and interaction tests between SGV status and treatment were performed. RESULTS Of 762 participants, 29% harbored an SGV. The SGV mutant group had worse OS (hazard ratio [HR], 1.28, 95% CI, 1.06 to 1.54), and within each treatment arm (docetaxel: HR, 1.31; atezolizumab: HR, 1.27). In the atezolizumab arm, the SGV mutant group compared with wild-type had worse OS in the PD-L1 high (HR, 1.31 [95% CI, 0.59 to 2.91]) and low (HR, 1.38 [95% CI, 0.98 to 1.93]) subgroups. SGV with missense, splice, and nonsense mutations had significantly worse OS than wild-type in the docetaxel arm (log-rank P = .01) but not in the atezolizumab arm (log-rank P = .33). SGV status did not predict greater OS benefit with atezolizumab over docetaxel (interaction P = .67). CONCLUSION In advanced NSCLC after chemotherapy progression, plasma-detected SGVs are common, and associated with inferior OS. Plasma SGV status should be considered as a stratification factor in future trials.
Collapse
Affiliation(s)
- Deborah Di-Xin Zhou
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| | | | | | - Frank Po-Yen Lin
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW Health Pathology, Camperdown, NSW, Australia
- Faculty of Medicine and Health and Western Sydney University School of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Milita Zaheed
- Prince of Wales Hereditary Cancer Centre, Randwick, NSW, Australia
| | - Robert John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
- St George Hospital, Kogarah, NSW, Australia
| |
Collapse
|
7
|
Li Y, Chen D, Xu Y, Ding Q, Xu X, Li Y, Mi Y, Chen Y. Prognostic implications, genomic and immune characteristics of lung adenocarcinoma with lepidic growth pattern. J Clin Pathol 2025; 78:277-284. [PMID: 39097406 DOI: 10.1136/jcp-2024-209603] [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: 04/24/2024] [Accepted: 07/17/2024] [Indexed: 08/05/2024]
Abstract
AIMS Conflicting data were provided regarding the prognostic impact and genomic features of lung adenocarcinoma (LUAD) with lepidic growth pattern (LP+A). Delineation of the genomic and immune characteristics of LP+A could provide deeper insights into its prognostic implications and treatment determination. METHODS We conducted a search of articles in PubMed, EMBASE and the Cochrane Library from inception to January 2024. A domestic cohort consisting of 52 LUAD samples was subjected to whole-exome sequencing as internal validation. Data from The Cancer Genomic Atlas and the Gene Expression Omnibus datasets were obtained to characterise the genomic and immune profiles of LP+A. Pooled HRs and rates were calculated. RESULTS The pooled results indicated that lepidic growth pattern was either predominant (0.35, 95% CI 0.22 to 0.56, p<0.01) or minor (HR 0.50, 95% CI 0.36 to 0.70, p<0.01) histological subtype was associated with favourable disease-free survival. Pooled gene mutation rates suggested higher EGFR mutation (0.55, 95% CI 0.46 to 0.64, p<0.01) and lower KRAS mutation (0.14, 95% CI 0.02 to 0.25, p=0.02) in lepidic-predominant LUAD. Lepidic-predominant LUAD had lower tumour mutation burden and pooled positive rate of PD-L1 expression compared with other subtypes. LP+A was characterised by abundance in resting CD4+memory T cells, monocytes and γδ T cells, as well as scarcity of cancer-associated fibroblasts. CONCLUSIONS LP+A was a unique histological subtype with a higher EGFR mutation rate, lower tumour mutation burden and immune checkpoint expression levels. Our findings suggested potential benefits from targeted therapy over immunotherapy in LP+A.
Collapse
Affiliation(s)
- Yue Li
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Donglai Chen
- Department of Thoracic Surgery, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Yi Xu
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qifeng Ding
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuejun Xu
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yongzhong Li
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yedong Mi
- Department of Thoracic Surgery, Jiangyin People's Hospital, Jiangyin, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
8
|
Song X, Pan Z, Zhang Y, Yang W, Zhang T, Wang H, Chen Y, Yu X, Ding H, Li R, Ge P, Xu L, Dong G, Jiang F. Excessive MYC Orchestrates Macrophages induced Chromatin Remodeling to Sustain Micropapillary-Patterned Malignancy in Lung Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403851. [PMID: 39899538 PMCID: PMC11948069 DOI: 10.1002/advs.202403851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 01/15/2025] [Indexed: 02/05/2025]
Abstract
Current understanding of micropapillary (MP)-subtype lung adenocarcinoma (LUAD) remains confined to biological activities and genomic landscapes. Unraveling the major regulatory programs underlying MP patterned malignancy offers opportunities to identify more feasible therapeutic targets for patients with MP LUAD. This study shows that patients with MP subtype LUAD have aberrant activation of the MYC pathway compared to patients with other subtypes. In vitro and xenograft mouse model studies reveal that MP pattern in malignancy cannot be solely due to aberrant MYC expression but requires the involvement of M2-like macrophages. Excessively expressed MYC leads to the accumulation of M2-like macrophages from the bone marrow, which secretes TGFβ, to induce the expression of FOSL2 in tumor cells, thereby remodeling chromatin accessibility at promoter regions of MP-pattern genes to promote the MYC-mediated de novo transcriptional regulation of these genes. Additionally, the MP-pattern in malignancy can be effectively alleviated by disrupting the TGFβ-FOSL2 axis. These findings reveal new functions for the M2-like macrophage-TGFβ-FOSL2 axis in MYC-overexpressing MP-subtype LUAD, identifying targetable vulnerabilities in this pathway.
Collapse
Affiliation(s)
- Xuming Song
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Zehao Pan
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
| | - Wenmin Yang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Drum Tower hospitalNanjing210008P.R. China
| | - Te Zhang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Biochemistry and Molecular GeneticsFeinberg School of MedicineNorthwestern UniversityChicagoIllinois60201USA
| | - Hui Wang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yuzhong Chen
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Xinnian Yu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Hanlin Ding
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Rutao Li
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow UniversityNanjing215000P. R. China
| | - Pengfei Ge
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Lin Xu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjing211116P. R. China
| | - Gaochao Dong
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| | - Feng Jiang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| |
Collapse
|
9
|
Matsuura Y, Onuma K, Coppo R, Uematsu H, Kondo J, Miyagawa‐Hayashino A, Takeda‐Miyata N, Kameyama K, Furuya T, Okada S, Shimomura M, Inoue M, Inoue M. Dynamic change of polarity in spread through air spaces of pulmonary malignancies. J Pathol 2025; 265:260-273. [PMID: 39804150 PMCID: PMC11794978 DOI: 10.1002/path.6382] [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: 04/04/2024] [Revised: 10/16/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025]
Abstract
Spread through air spaces (STAS) is a histological finding of lung tumours where tumour cells exist within the air space of the lung parenchyma beyond the margin of the main tumour. Although STAS is an important prognostic factor, the pathobiology of STAS remains unclear. Here, we investigated the mechanism of STAS by analysing the relationship between STAS and polarity switching in vivo and in vitro. Histopathological analysis revealed that apical membranes were observed outside the STAS lesions around colorectal cancer (CRC) lung metastases and lung adenocarcinomas. When apical-out CRC organoids were administered intratracheally to mice, the organoids had greater metastatic potential than did single cells. To investigate the pathobiology of STAS, we established an in vitro model of STAS in which CRC or lung cancer organoids were co-cultured with 2D-cultured mouse airway epithelial organoids (2D-MAOs). Adhesion of cancer organoids to 2D-MAOs was much less than to type I collagen or endothelial cells, suggesting a protective role of the airway epithelium against adhesion. Loss of the apical membrane of CRC organoids at the contact surface with 2D-MAOs after adhesion was responsible for establishing adhesion. When airway epithelium was stimulated by transforming growth factor beta 1 (TGF-β1), adhesion of CRC organoids was enhanced. Among TGF-β1-induced genes in airway epithelium, follistatin-like protein 1 (FSTL1) increased CRC organoid adhesion by promoting loss of the apical membrane. These results suggested that TGF-β1-induced FSTL1 may promote metastatic progression of STAS by altering the polarity status. Elucidating the mechanism of STAS could contribute to the improvement of survival in patients with pulmonary malignancies associated with STAS. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Grants
- 21cm0106203h0006 Japan Agency for Medical Research and Development
- 18H02648 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 20H03772 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 20K08286 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 22K08982 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- 23K07395 Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology in Japan
- Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Yoshiaki Matsuura
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kunishige Onuma
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroyuki Uematsu
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- KBBM Inc.KyotoJapan
| | - Jumpei Kondo
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Department of Molecular Biology and Clinical Investigation, Graduate School of MedicineOsaka UniversityOsakaJapan
| | | | - Naoko Takeda‐Miyata
- Department of Surgical PathologyKyoto Prefectural University of MedicineKyotoJapan
| | - Kenji Kameyama
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Tatsuo Furuya
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Satoru Okada
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masanori Shimomura
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masayoshi Inoue
- Divison of Thoracic Surgery, Department of Surgery, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Inoue
- Department of Clinical Bio‐resource Research and Development, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
10
|
Xin S, Wen M, Tian Y, Dong H, Wan Z, Jiang S, Meng F, Xiong Y, Han Y. Impact of histopathological subtypes on invasive lung adenocarcinoma: from epidemiology to tumour microenvironment to therapeutic strategies. World J Surg Oncol 2025; 23:66. [PMID: 40016762 PMCID: PMC11866629 DOI: 10.1186/s12957-025-03701-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/02/2025] [Indexed: 03/01/2025] Open
Abstract
Lung adenocarcinoma is the most prevalent type of lung cancer, with invasive lung adenocarcinoma being the most common subtype. Screening and early treatment of high-risk individuals have improved survival; however, significant differences in prognosis still exist among patients at the same stage, especially in the early stages. Invasive lung adenocarcinoma has different histological morphologies and biological characteristics that can distinguish its prognosis. Notably, several studies have found that the pathological subtypes of invasive lung adenocarcinoma are closely associated with clinical treatment. This review summarised the distribution of various pathological subtypes of invasive lung adenocarcinoma in the population and their relationship with sex, smoking, imaging features, and other histological characteristics. We comprehensively analysed the genetic characteristics and biomarkers of the different pathological subtypes of invasive lung adenocarcinoma. Understanding the interaction between the pathological subtypes of invasive lung adenocarcinoma and the tumour microenvironment helps to reveal new therapeutic targets for lung adenocarcinoma. We also extensively reviewed the prognosis of various pathological subtypes and their effects on selecting surgical methods and adjuvant therapy and explored future treatment strategies.
Collapse
Affiliation(s)
- Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- Department of Thoracic Surgery, 962 Hospital of the Joint Logistics Support Force, Harbin, China
| | - Miaomiao Wen
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Honghong Dong
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Zitong Wan
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- College of Life Sciences, Northwestern University, Xi'an, 710069, China
| | - Suxin Jiang
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Fancheng Meng
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Innovation Center for Advanced Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
- Department of Thoracic Surgery, First Medical Center, Chinese PLA General Hospital and PLA Medical School, Beijing, China.
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Road, Baqiao District, Shaanxi, , Xi'an, 710038, China.
| | - Yong Han
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, Beijing, China.
- Department of Thoracic Surgery, Air Force Medical Center, Fourth Military Medical University, 30 Fucheng Road, Haidian District, Shaanxi, , Beijing, 100142, China.
| |
Collapse
|
11
|
Driussi A, Lamaze FC, Kordahi M, Armero VS, Gaudreault N, Orain M, Enlow W, Abbosh C, Hodgson D, Dasgupta A, Gagné A, Bossé Y, Joubert P. Clinicopathological Predictors of the Presence of Blood Circulating Tumor DNA in Early-Stage Non-Small Cell Lung Cancers. Mod Pathol 2025; 38:100744. [PMID: 40020968 DOI: 10.1016/j.modpat.2025.100744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
The implementation of lung cancer screening programs across the world has drawn considerable attention to improving early-stage lung cancer detection and prognostication. Several blood-based assays detecting circulating tumor DNA (ctDNA) recently emerged as noninvasive methods to detect malignancies. However, their limited sensitivity and predictive value remain a hurdle to their clinical use. We aimed to evaluate the association between clinicopathological parameters and presurgical ctDNA detection in clinical stage I non-small cell lung cancer patients to further understand ctDNA shedding biology. The cohort included 180 adenocarcinomas (LUAD) and 80 squamous cell carcinomas (LUSC) stage I patients who underwent lung cancer resection. Patients' clinical and pathological features were collected. A multicancer early-detection test (GRAIL LLC) was used to detect ctDNA using targeted methylation patterns. The association between the cell-free DNA tumor methylated fraction (TMeF) and the clinicopathological predictors was evaluated using univariate and multivariate modeling. LUSC was associated with a higher TMeF than LUAD. Pathological stage, tumor grade, and tumor volume were key determinants of ctDNA detection in both LUSC and LUAD. In LUAD, ctDNA detection also correlated with histologic pattern composition, necrosis, acute inflammation, and, to a lesser degree, spread through alveolar spaces and lymphovascular invasion. Based on our results, we propose classification methods for both LUAD (using histologic pattern composition) and LUSC (using tumor grade and pathological stage) to identify patients likely to have high ctDNA levels. These results confirm previous findings and suggest that previously unidentified factors, including histologic pattern composition and acute inflammation, influence ctDNA levels. These results will help in understanding the ctDNA shedding process and may allow identification of patients eligible for ctDNA detection-based follow-up.
Collapse
Affiliation(s)
- Arnaud Driussi
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Fabien C Lamaze
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Manal Kordahi
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Victoria Saavedra Armero
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Michèle Orain
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - William Enlow
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Chris Abbosh
- Translational Medicine Early Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Darren Hodgson
- Translational Medicine Early Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Abhijit Dasgupta
- Oncology Data Science, Oncology R&D, AstraZeneca, Gaithersburg, Maryland
| | - Andréanne Gagné
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada; Department of Molecular Medicine, Université Laval, Quebec City, Canada
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada; Department of Molecular Biology, Pathology and Medical Biochemistry, Université Laval, Quebec City, Canada.
| |
Collapse
|
12
|
Li Z, Chen L, Wei Z, Liu H, Zhang L, Huang F, Wen X, Tian Y. A novel classification method for LUAD that guides personalized immunotherapy on the basis of the cross-talk of coagulation- and macrophage-related genes. Front Immunol 2025; 16:1518102. [PMID: 40018029 PMCID: PMC11866059 DOI: 10.3389/fimmu.2025.1518102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Purpose The coagulation process and infiltration of macrophages affect the progression and prognosis of lung adenocarcinoma (LUAD) patients. This study was designed to explore novel classification methods that better guide the precise treatment of LUAD patients on the basis of coagulation and macrophages. Methods Weighted gene coexpression network analysis (WGCNA) was applied to identify M2 macrophage-related genes, and TAM marker genes were acquired through the analysis of scRNA-seq data. The MSigDB and KEGG databases were used to obtain coagulation-associated genes. The intersecting genes were defined as coagulation and macrophage-related (COMAR) genes. Unsupervised clustering analysis was used to evaluate distinct COMAR patterns for LUAD patients on the basis of the COMAR genes. The R package "limma" was used to identify differentially expressed genes (DEGs) between COMAR patterns. A prognostic risk score model, which was validated through external data cohorts and clinical samples, was constructed on the basis of the COMAR DEGs. Results In total, 33 COMAR genes were obtained, and three COMAR LUAD subtypes were identified on the basis of the 33 COMAR genes. There were 341 DEGs identified between the three COMAR subtypes, and 60 prognostic genes were selected for constructing the COMAR risk score model. Finally, 15 prognosis-associated genes (CORO1A, EPHA4, FOXM1, HLF, IFIH1, KYNU, LY6D, MUC16, PPARG, S100A8, SPINK1, SPINK5, SPP1, VSIG4, and XIST) were included in the model, which was efficient and robust in predicting LUAD patient prognosis and clinical outcomes in patients receiving anti-PD-1/PD-L1 immunotherapy. Conclusions LUAD can be classified into three subtypes according to COMAR genes, which may provide guidance for precise treatment.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ling Chen
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Hongtao Liu
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Clinical Pathology, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, China
| | - Lu Zhang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fujing Huang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Wen
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Tian
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L, Li W. Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 2025; 16:1477. [PMID: 39929832 PMCID: PMC11811181 DOI: 10.1038/s41467-024-55068-2] [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: 06/27/2022] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Post-operative recurrence rates of stage I non-small cell lung cancer (NSCLC) range from 20% to 40%. Nonetheless, the molecular mechanisms underlying recurrence hitherto remain largely elusive. Here, we generate genomic, epigenomic and transcriptomic profiles of paired tumors and adjacent tissues from 122 stage I NSCLC patients, among which 57 patients develop recurrence after surgery during follow-up. Integrated analyses illustrate that the presence of predominantly solid or micropapillary histological subtypes, increased genomic instability, and APOBEC-related signature are associated with recurrence. Furthermore, TP53 missense mutation in DNA-binding domain could contribute to shorter time to recurrence. DNA hypomethylation is pronounced in recurrent NSCLC, and PRAME is the significantly hypomethylated and overexpressed gene in recurrent lung adenocarcinoma (LUAD). Mechanistically, hypomethylation at TEAD1 binding site facilitates the transcriptional activation of PRAME. Inhibition of PRAME restrains the tumor metastasis via downregulation of epithelial-mesenchymal transition-related genes. We also identify that enrichment of AT2 cells with higher copy number variation burden, exhausted CD8 + T cells and Macro_SPP1, along with the reduced interaction between AT2 and immune cells, is essential for the formation of ecosystem in recurrent LUAD. Finally, multi-omics clustering could stratify the NSCLC patients into 4 subclusters with varying recurrence risk and subcluster-specific therapeutic vulnerabilities. Collectively, this study constitutes a promising resource enabling insights into the biological mechanisms and clinical management for post-operative recurrence of stage I NSCLC.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyao Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lujia Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changshu Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Qiu
- Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Li Z, Lin X, Yang Y, Tian M, Zhang L, Huang F, Wen X, Wei Z, Tian Y. EXO1 is a key gene for lung-resident memory T cells and has diagnostic and predictive values for lung adenocarcinoma. Sci Rep 2025; 15:4002. [PMID: 39893221 PMCID: PMC11787328 DOI: 10.1038/s41598-025-88126-w] [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: 10/05/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a very common and lethal kind of lung malignancy. An increasing number of studies indicated that tissue-resident memory T (TRM) cells played significant roles in anti-cancer immunity. In our previous study, EXO1 was found to be a core gene for TRM cells in the prognosis of LUAD. However, the roles of EXO1 in the tumor microenvironment, and its application in the diagnosis and prognosis prediction of LUAD are still inadequately explored. In this study, the RNA expression, DNA methylation, CNV, somatic mutation data of EXO1, and the corresponding patients' clinical information from publicly available databases were analyzed using bioinformatic methods. The results were validated through immunohistochemical staining of EXO1 in LUAD samples. The results showed EXO1 was aberrantly highly expressed in LUAD tissues. High expression of EXO1 was a risky factor for LUAD patients. The expression level of EXO1 was associated with many clinical features such as TNM stages. It can also distinguish normal tissues and LUAD tumor tissues accurately. EXO1 expression was correlated with the infiltration of immune cells, and high expression of EXO1 was an adverse effect on LUAD patients receiving anti-PD-1/PD-L1 immunotherapy. Moreover, patients with EXO1 mutation had worse DSS, DFI and PFI.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital, Shandong University, 250021, Jinan, P.R. China
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, P.R. China
| | - Yuanhui Yang
- Department of Pathology, Shandong Provincial Hospital, Shandong University, 250021, Jinan, P.R. China
| | - Mei Tian
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, P.R. China
| | - Lu Zhang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Fujing Huang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Xiao Wen
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, 250014, Jinan, P.R. China.
| | - Yuan Tian
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250011, Jinan, P.R. China.
| |
Collapse
|
15
|
Jiang W, Zhang F, Tang Z, Xu S, Zhang Y, Liu L, Zhong D, Liu Y. Prediction of prognosis and immune response in lung adenocarcinoma based on mitophagy and lactate-related gene signatures. Int J Clin Oncol 2025; 30:277-297. [PMID: 39601968 DOI: 10.1007/s10147-024-02664-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) causes leading death worldwide. Mitophagy and lactate metabolism accumulation are distinctive features of LUAD. We aimed to identify lactate-related genes (LRGs) signatures based on mitophagy for predicting prognosis and immune response in LUAD. METHODS The gene expression and clinical data were downloaded from TCGA and GEO database. First, the subtype analysis was analyzed based on 29 mitophagy genes. Survival, immune, and function differences between the different subtypes were analyzed. Then, based on mitophagy genes and 14 LRGs, the best LRGs were screened to construct a risk score model and combined with clinical factors to establish a nomogram for predicting patient survival. Finally, the expression level and molecular function of the key candidate gene OGDH were verified by in vitro experiments. RESULTS All the LUAD samples were divided into 2 subtypes: sub1 and sub2. The sub2 possessed worse survival. Immune score, immune checkpoint genes, and human leucocyte antigen genes in sub1 were higher than in sub2. Six optimal mitophagy-related LRGs were used to construct a risk score model. A high-risk score indicates poorer survival, higher tumor mutation burden, and higher drug sensitivity. The nomogram was robust in predicting LUAD survival. The experiments in vitro showed that knockdown of OGDH inhibited the proliferation, migration and invasion in LUAD cells. CONCLUSIONS A nomogram based on the construction of the mitophagy-related lactate genes predicts prognosis and immune response in LUAD. These results could help with risk stratification and targeted therapy for LUAD.
Collapse
Affiliation(s)
- Wenjie Jiang
- Department of Thoracic Surgery, Tangdu Hospital, the Air Force Medical University, Xi'an, 710038, China
- Department of Thoracic Surgery, Weinan Central Hospital, Weinan, 714000, China
| | - Fan Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zhen Tang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Shuonan Xu
- Department of Thoracic Surgery, Tangdu Hospital, the Air Force Medical University, Xi'an, 710038, China
| | - Yukun Zhang
- Department of Cardiology, Tangdu Hospital, The Air Force Medical University, Xi'an 710038, China
| | - Lina Liu
- Department of Clinical Laboratory, Xi'an Peoples' Hospital (Xi'an Fourth Hospital), Xi'an, 710004, Shaanxi, China.
| | - Daixing Zhong
- Department of Thoracic Surgery, Tangdu Hospital, the Air Force Medical University, Xi'an, 710038, China.
| | - Yingxiang Liu
- Department of Orthopedic Surgery, Orthopedic Oncology Institute, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China.
| |
Collapse
|
16
|
Passarella G, Canova S, Abbate MI, Caspani G, Sala L, Russo A, Muscolino P, Colonese F, Cortinovis DL. The hype around ctDNA guiding an informed perioperative therapeutic strategy in early-stage non-small cell lung cancer. Discov Oncol 2025; 16:100. [PMID: 39881042 PMCID: PMC11780067 DOI: 10.1007/s12672-025-01826-7] [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: 10/11/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a dire disease being the first cause of cancer death among both genders. Early-stage NSCLC often has better treatment outcomes despite it being a highly heterogeneous disease. So far, the neo-adjuvant chemotherapy strategies have led to a small benefit with an improvement of 5% in overall survival as an absolute benefit. Recently, the introduction of immune checkpoint inhibitors combined with chemotherapy has shown robust efficacy in terms of event-free survival and overall survival. Thus, these combinations are today considered a new standard of care in early-stage NSCLC. The application of these strategies to all-comer population lead to confounding definitive results regarding the efficacy and predictive biomarkers are urgently needed balancing the promise of healing than toxicities. At present, the clinical staging TNM system guides the clinical choice, however it is not entirely sufficient. Circulant tumoral DNA (ctDNA) emerged as a promising prognostic and predictive biomarker that may guide the future perioperative strategy and pave the way to personalized medicine also in this exciting field. This narrative review aims to put in the context the employment of ctDNA, give some perspective and suggestions weighing the pros and cons of this technique for our tomorrow clinical practice.
Collapse
Affiliation(s)
- Gaia Passarella
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Medicine and Surgery Department, University of Brescia, Brescia, Italy.
| | - Stefania Canova
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| | - Maria Ida Abbate
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Caspani
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy
| | - Luca Sala
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandro Russo
- Department of Medical Oncology, Humanitas Istituto Clinico Catanese, Misterbianco, Catania, Italy
| | - Paola Muscolino
- Department of Human Pathology "G. Barresi", Medical Oncology Specialization School, University of Messina, Messina, Italy
| | - Francesca Colonese
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Diego Luigi Cortinovis
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy
| |
Collapse
|
17
|
Black JRM, Bartha G, Abbott CW, Boyle SM, Karasaki T, Li B, Chen R, Harris J, Veeriah S, Colopi M, Bakir MA, Liu WK, Lyle J, Navarro FCP, Northcott J, Pyke RM, Hill MS, Thol K, Huebner A, Bailey C, Colliver EC, Martínez-Ruiz C, Grigoriadis K, Pawlik P, Moore DA, Marinelli D, Shutkever OG, Murphy C, Sivakumar M, Shaw JA, Hackshaw A, McGranahan N, Jamal-Hanjani M, Frankell AM, Chen RO, Swanton C. Ultrasensitive ctDNA detection for preoperative disease stratification in early-stage lung adenocarcinoma. Nat Med 2025; 31:70-76. [PMID: 39806071 PMCID: PMC11750713 DOI: 10.1038/s41591-024-03216-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 07/29/2024] [Indexed: 01/16/2025]
Abstract
Circulating tumor DNA (ctDNA) detection can predict clinical risk in early-stage tumors. However, clinical applications are constrained by the sensitivity of clinically validated ctDNA detection approaches. NeXT Personal is a whole-genome-based, tumor-informed platform that has been analytically validated for ultrasensitive ctDNA detection at 1-3 ppm of ctDNA with 99.9% specificity. Through an analysis of 171 patients with early-stage lung cancer from the TRACERx study, we detected ctDNA pre-operatively within 81% of patients with lung adenocarcinoma (LUAD), including 53% of those with pathological TNM (pTNM) stage I disease. ctDNA predicted worse clinical outcome, and patients with LUAD with <80 ppm preoperative ctDNA levels (the 95% limit of detection of a ctDNA detection approach previously published in TRACERx) experienced reduced overall survival compared with ctDNA-negative patients with LUAD. Although prospective studies are needed to confirm the clinical utility of the assay, these data show that our approach has the potential to improve disease stratification in early-stage LUADs.
Collapse
Affiliation(s)
- James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | | | | | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | | | - Rui Chen
- Personalis Inc., Fremont, CA, USA
| | | | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Martina Colopi
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Wing Kin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | | | | | | | - Mark S Hill
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Chris Bailey
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Emma C Colliver
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kristiana Grigoriadis
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Piotr Pawlik
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Daniele Marinelli
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Oliver G Shutkever
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Cian Murphy
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Jacqui A Shaw
- Leicester NIHR BRC & University of Leicester, Leicester, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
18
|
Thunnissen E, Blaauwgeers H, Filipello F, Lissenberg-Witte B, Minami Y, Noguchi M, Quesne JL, Papotti MG, Flieder DB, Pelosi G, Sansano I, Berezowska S, Ryška A, Brcic L, Motoi N, Nakatani Y, Kuempers C, Hofman P, Hofman V, Dale VG, Rossi G, Ambrosi F, Matsubara D, Ishikawa Y, Weynand B, Calabrese F, Pezzuto F, Kern I, Nicholson S, Mutka A, Dacic S, Beasley MB, Arrigoni G, Timens W, Ooft M, Brinkhuis M, Bulkmans N, Britstra R, Vreuls W, Jones KD, von der Thüsen JH, Hager H, Perner S, Moore D, Leonte DG, Al-Janabi S, Schønau A, Neumann O, Kluck K, Ourailidis I, Ball M, Budczies J, Kazdal D, Stenzinger A. A reproducibility study on invasion in small pulmonary adenocarcinoma according to the WHO and a modified classification, supported by biomarkers. Lung Cancer 2025; 199:108060. [PMID: 39793325 DOI: 10.1016/j.lungcan.2024.108060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVES Evaluating invasion in non-mucinous adenocarcinoma (NMA) of the lung is crucial for accurate pT-staging. This study compares the World Health Organization (WHO) with a recently modified NMA classification. MATERIALS AND METHODS A retrospective case-control study was conducted on small NMA pT1N0M0 cases with a 5-year follow-up. Seventy cases were reviewed by 42 pulmonary pathologists first according to the WHO classification and after tutorial according to a modified classification. A third round was conducted based on feedback from 41 peers of previous rounds. Additionally, orthogonal biomarker analysis was performed. RESULTS In the first two rounds, 42 pathologists from 13 countries assessed all 70 cases, while 36 pathologists evaluated 41 non-unanimous cases in the third round. Kappa values for invasiveness increased in rounds 1, 2, and 3 to 0.27, 0.45 and 0.62, respectively. In contrast to low variation in total tumor size measurements (6 %), a marked increase in invasive tumor size variation was observed (42 %), which was associated with high uncertainty. In the third round 10 cases were non-invasive, all without recurrence. The modified classification showed in the 3rd round marked reduction of the variation in pT staging compared to the current WHO classification. Proliferation rate, tumor mutational burden, and transcriptomic profiles supported the distinction between invasive cases and non-invasive cases of the modified classification. CONCLUSION The modified classification demonstrates essentially higher reproducibility compared to the current WHO classification in NMA. The modified classification proves valuable in identifying low-risk lesions that are entirely non-invasive, and is supported by biomarker analysis.
Collapse
Affiliation(s)
- Erik Thunnissen
- Dept. of Pathology, Amsterdam UMC, VU University, Amsterdam, the Netherlands.
| | | | | | - Birgit Lissenberg-Witte
- Dept. of Epidemiology and Data Science, Amsterdam UMC, VU University, Amsterdam, the Netherlands
| | - Yuko Minami
- Dept. of Pathology, National Hospital Organization Ibarakihigashi National Hospital, Tokai, Japan
| | - Masayuki Noguchi
- Dept. of Pathology, Naritatomisato Tokushukai Hospital, Chiba, Japan
| | - John Le Quesne
- Dept. of Pathology, School of Cancer Sciences, University of Glasgow, Scotland, UK; Dept. of Pathology, CRUK Beatson Cancer Research Institute, Glasgow, Scotland, UK; Dept. of Pathology, Department of Histopathology, Queen Elizabeth University Hospital, Glasgow, Scotland, UK
| | | | | | - Giuseppe Pelosi
- Dept. of Pathology, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Irene Sansano
- Dept. of Pathology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Sabina Berezowska
- Dept. of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Aleš Ryška
- Dept. of Pathology, Charles University, ESP, Hradec Kralove, Czech Republic
| | - Luka Brcic
- Dept. of Pathology, Medical University of Graz, Graz, Austria
| | - Noriko Motoi
- Dept. of Pathology, Saitama Cancer Center, Saitama, Japan
| | - Yukio Nakatani
- Dept. of Pathology, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Christiane Kuempers
- Dept. of Pathology, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Paul Hofman
- IHU RespirERA, FHU OncoAge, Nice University Hospital Center, Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Veronique Hofman
- IHU RespirERA, FHU OncoAge, Nice University Hospital Center, Laboratory of Clinical and Experimental Pathology, Nice, France
| | - Vibeke Grotnes Dale
- Dept. of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Norwegian University of Science and Technology, Trondheim, Norway
| | - Giulio Rossi
- Dept. of Pathology, Fondazione Poliambulanza Hospital Institute, Brescia, Brescia, Italy
| | - Francesca Ambrosi
- Dept. of Pathology, Maggiore Hospital, University of Bologna, Bologna, Italy
| | | | - Yuichi Ishikawa
- Dept. of Pathology, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | | | | | | | - Izidor Kern
- Dept. of Pathology, St. James's Hospital, Dublin, Ireland
| | - Siobhan Nicholson
- Dept. of Pathology, HUS Helsinki University Hospital, Helsinki, Finland
| | - Aino Mutka
- Dept. of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Sanja Dacic
- Dept. of Pathology, Mount Sinai Medical Center, New York, NY, USA
| | - Mary Beth Beasley
- Dept. of Pathology, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Wim Timens
- Dept. of Pathology, Rijnstate Ziekenhuis, Arnhem, the Netherlands
| | - Marc Ooft
- Dept. of Pathology, LabPON, Hengelo, the Netherlands
| | - Mariel Brinkhuis
- Dept. Pathologie-DNA, St. Antoniusziekenhuis, Nieuwegein, the Netherlands
| | - Nicole Bulkmans
- Dept. of Pathology, Meander Medisch Centrum, Amersfoort, the Netherlands
| | - Rieneke Britstra
- Dept. of Pathology, Canisius Wilhelmina Hospital, Nijmegen, the Netherlands
| | - Willem Vreuls
- Dept. of Pathology, University of California, San Francisco, CA, USA
| | - Kirk D Jones
- Dept. of Pathology, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Hendrik Hager
- Dept. of Pathology, University College London Cancer Institute, London, United Kingdom
| | - Sven Perner
- Dept. of Pathology, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - David Moore
- Dept. of Pathology, University of California, San Francisco, CA, USA
| | | | | | | | - Olaf Neumann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Iordanis Ourailidis
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Markus Ball
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Center for Personalized Medicine (ZPM) Heidelberg, Heidelberg, Germany; Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
19
|
Xu J, Liu L, Ji Y, Yan T, Shi Z, Pan H, Wang S, Yu K, Qin C, Zhang T. Enhanced CT-Based Intratumoral and Peritumoral Radiomics Nomograms Predict High-Grade Patterns of Invasive Lung Adenocarcinoma. Acad Radiol 2025; 32:482-492. [PMID: 39095263 DOI: 10.1016/j.acra.2024.07.026] [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: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
RATIONALE AND OBJECTIVES Extraction of intratumoral and peritumoral radiomics features combined with clinical factors to establish nomograms to predict high-grade patterns (micropapillary and solid) of invasive adenocarcinoma of the lung (IAC). MATERIALS AND METHODS A retrospective study was conducted on 463 patients with pathologically confirmed IAC. Patients were randomized in a 7:3 ratio into a training cohort (n = 324) and a testing cohort (n = 139). A total of 2154 CT-based radiomic features were extracted from each of the four regions: gross tumor volume (GTV) and gross peritumoral tumor volume (GPTV3, GPTV6, GPTV9) containing peri-tumor regions of 3 mm, 6 mm, and 9 mm. A radiomics nomogram was constructed based on the optimal radiomics model and clinically independent predictors. RESULTS The GPTV3 radiomics model showed better predictive performance in the testing group compared to the GTV (0.840), GPTV6 (0.843), and GPTV9 (0.734) models, with an AUC value of 0.889 in the testing group. In the clinical model, tumor density and the presence of a spiculation sign were identified as independent predictors. The nomogram, which combined these independent predictors with the GPTV3-Radscore, proved to be clinically useful. CONCLUSION The GPTV3 radiomics model was superior to the GTV, GPTV6, and GPTV9 radiomics models in predicting high-grade patterns (HGP) of IAC. In addition, nomograms based on GPTV3 radiomics features and clinically independent predictors can further improve the prediction efficiency.
Collapse
Affiliation(s)
- Jiaheng Xu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ling Liu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Ji
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tiancai Yan
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhenzhou Shi
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hong Pan
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuting Wang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kang Yu
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chunhui Qin
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tong Zhang
- Department of Radiology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
20
|
Biswas D, Liu YH, Herrero J, Wu Y, Moore DA, Karasaki T, Grigoriadis K, Lu WT, Veeriah S, Naceur-Lombardelli C, Magno N, Ward S, Frankell AM, Hill MS, Colliver E, de Carné Trécesson S, East P, Malhi A, Snell DM, O'Neill O, Leonce D, Mattsson J, Lindberg A, Micke P, Moldvay J, Megyesfalvi Z, Dome B, Fillinger J, Nicod J, Downward J, Szallasi Z, Hackshaw A, Jamal-Hanjani M, Kanu N, Birkbak NJ, Swanton C. Prospective validation of ORACLE, a clonal expression biomarker associated with survival of patients with lung adenocarcinoma. NATURE CANCER 2025; 6:86-101. [PMID: 39789179 PMCID: PMC11779643 DOI: 10.1038/s43018-024-00883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/15/2024] [Indexed: 01/12/2025]
Abstract
Human tumors are diverse in their natural history and response to treatment, which in part results from genetic and transcriptomic heterogeneity. In clinical practice, single-site needle biopsies are used to sample this diversity, but cancer biomarkers may be confounded by spatiogenomic heterogeneity within individual tumors. Here we investigate clonally expressed genes as a solution to the sampling bias problem by analyzing multiregion whole-exome and RNA sequencing data for 450 tumor regions from 184 patients with lung adenocarcinoma in the TRACERx study. We prospectively validate the survival association of a clonal expression biomarker, Outcome Risk Associated Clonal Lung Expression (ORACLE), in combination with clinicopathological risk factors, and in stage I disease. We expand our mechanistic understanding, discovering that clonal transcriptional signals are detectable before tissue invasion, act as a molecular fingerprint for lethal metastatic clones and predict chemotherapy sensitivity. Lastly, we find that ORACLE summarizes the prognostic information encoded by genetic evolutionary measures, including chromosomal instability, as a concise 23-transcript assay.
Collapse
Grants
- C11496/A17786, C416/A21999 Cancer Research UK (CRUK)
- CC2041 Wellcome Trust
- CC2041 Arthritis Research UK
- Young Investigator Grant International Association for the Study of Lung Cancer (IASLC)
- 202060447 Japan Society for the Promotion of Science London (JSPS London)
- I4677 Austrian Science Fund (Fonds zur Förderung der Wissenschaftlichen Forschung)
- Wellcome Trust
- 220589/Z/20/Z Wellcome Trust (Wellcome)
- EDDCPJT\100008 Cancer Research UK (CRUK)
- UCL/12/0279 University College London (UCL)
- Bolyai Research Scholarship Hungarian Academy of Sciences | Magyar Tudományos Akadémia Számítástechnikai és Automatizálási Kutatóintézet (Számítástechnikai és Automatizálási Kutatóintézet)
- ID16584 Novo Nordisk Foundation Center for Basic Metabolic Research (NovoNordisk Foundation Center for Basic Metabolic Research)
- CTUQQR-DEC22/100009 Cancer Research UK
- Francis Crick Institute (Francis Crick Institute Limited)
- RCUK | Medical Research Council (MRC)
- Rosetrees Trust
- Breast Cancer Research Foundation (BCRF)
- Butterfield and Stoneygate Trusts National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre The Mark Foundation for Cancer Research Aspire Award (grant 21-029-ASP)
- Hungarian National Research, Development and Innovation Office (K129065)
- Hungarian National Research, Development, and Innovation Office (2020‐1.1.6‐JÖVŐ, TKP2021‐EGA‐33, FK‐143751 and FK-147045) New National Excellence Program of the Ministry for Innovation and Technology of Hungary (UNKP‐20‐3, UNKP‐21‐3 and UNKP-23-5)
- Lung Cancer Research Foundation (LCRF)
- DH | NIHR | Health Services Research Programme (NIHR Health Services Research Programme)
- NIH National Cancer Institute UKI NETs
- Hungarian National Research, Development, and Innovation Office (2020‐1.1.6‐JÖVŐ, TKP2021‐EGA‐33, FK‐143751 and FK-147045)
Collapse
Affiliation(s)
- Dhruva Biswas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
| | - Yun-Hsin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Javier Herrero
- Bill Lyons Informatics Centre, University College London Cancer Institute, London, UK
| | - Yin Wu
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, UK
- Department of Medical Oncology, Guy's Hospital, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Metastasis Lab, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | - Kristiana Grigoriadis
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Wei-Ting Lu
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Neil Magno
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Philip East
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Aman Malhi
- Cancer Research UK and University College London Cancer Trials Centre, University College London, London, UK
| | - Daniel M Snell
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Olga O'Neill
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Daniel Leonce
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Johanna Mattsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Amanda Lindberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Judit Moldvay
- 1st Department of Pulmonology, National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Pulmonology, University of Szeged Albert Szent-Gyorgyi Medical School, Szeged, Hungary
| | - Zsolt Megyesfalvi
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - János Fillinger
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Jerome Nicod
- Genomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Zoltan Szallasi
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Allan Hackshaw
- Cancer Research UK and University College London Cancer Trials Centre, University College London, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Lab, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
21
|
Xiao N, Liu H, Zhang C, Chen H, Li Y, Yang Y, Liu H, Wan J. Applications of single-cell analysis in immunotherapy for lung cancer: Current progress, new challenges and expectations. J Adv Res 2024:S2090-1232(24)00462-4. [PMID: 39401694 DOI: 10.1016/j.jare.2024.10.008] [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: 02/04/2024] [Revised: 06/28/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Lung cancer is a prevalent form of cancer worldwide, presenting a substantial risk to human well-being. Lung cancer is classified into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). The advancement of tumor immunotherapy, specifically immune checkpoint inhibitors and adaptive T-cell therapy, has encountered substantial obstacles due to the rapid progression of SCLC and the metastasis, recurrence, and drug resistance of NSCLC. These challenges are believed to stem from the tumor heterogeneity of lung cancer within the tumor microenvironment. AIM OF REVIEW This review aims to comprehensively explore recent strides in single-cell analysis, a robust sequencing technology, concerning its application in the realm of tumor immunotherapy for lung cancer. It has been effectively integrated with transcriptomics, epigenomics, genomics, and proteomics for various applications. Specifically, these techniques have proven valuable in mapping the transcriptional activity of tumor-infiltrating lymphocytes in patients with NSCLC, identifying circulating tumor cells, and elucidating the heterogeneity of the tumor microenvironment. KEY SCIENTIFIC CONCEPTS OF REVIEW The review emphasizes the paramount significance of single-cell analysis in mapping the immune cells within NSCLC patients, unveiling circulating tumor cells, and elucidating the tumor microenvironment heterogeneity. Notably, these advancements highlight the potential of single-cell analysis to revolutionize lung cancer immunotherapy by characterizing immune cell fates, improving therapeutic strategies, and identifying promising targets or prognostic biomarkers. It is potential to unravel the complexities within the tumor microenvironment and enhance treatment strategies marks a significant step towards more effective therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
22
|
Jensen SØ, Moore DA, Surani AA, Crosbie PAJ, Rosenfeld N, Rintoul RC. Second Primary Lung Cancer - An Emerging Issue in Lung Cancer Survivors. J Thorac Oncol 2024; 19:1415-1426. [PMID: 39059487 DOI: 10.1016/j.jtho.2024.07.014] [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: 03/07/2024] [Revised: 06/22/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
As a result of an increased focus on early detection including lung cancer screening, combined with more curative treatment options, the 5-year survival rates for lung cancer are improving. Welcome though this is, it brings new, hitherto unseen challenges. As more patients are cured and survive longer, they are at risk of developing second primary cancers, particularly lung cancer. In this review, we examine the challenges that surveillance, diagnosis, and management of second primary lung cancer (SPLC) bring and how these can be addressed. Recent data from prospective follow-up studies suggests that the incidence of SPLC may be higher than previously appreciated, partly due to an increase in multi-focal adenocarcinoma spectrum disease. Over 5 years, up to 1 in 6 long-term lung cancer survivors may develop a SPLC. Although not routinely used in clinical practice at present, genomic approaches for differentiating SPLC from intrapulmonary metastases of the first primary are emerging, and we highlight how this could be used to help differentiate lesions. An accurate distinction between SPLC and the recurrence of the first primary is of paramount importance due to the very different management strategies that may be required. Wrongly classifying an SPLC as a recurrence of the first primary may have significant consequences for patient management and overall survival. Updated approaches to the classification of SPLC combining clinical history, histopathological assessment, and genomic profiling are needed. Finally, we review the potential role of early detection biomarkers in the identification of SPLC, focusing in particular on blood-based biomarkers that are being examined in a multi-center prospective study recruiting lung cancer survivors.
Collapse
Affiliation(s)
- Sarah Østrup Jensen
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - David A Moore
- Department of Cellular Pathology, University College Hospital, London United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Arif A Surani
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Philip A J Crosbie
- Division of Immunology, Immunity and Infection and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Robert C Rintoul
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom; Department of Oncology, University of Cambridge, Cambridge, United Kingdom; Department of Thoracic Oncology, Royal Papworth Hospital, Cambridge, United Kingdom.
| |
Collapse
|
23
|
Zheng Y, Conrad RD, Green EJ, Burks EJ, Betke M, Beane JE, Kolachalama VB. Graph Attention-Based Fusion of Pathology Images and Gene Expression for Prediction of Cancer Survival. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:3085-3097. [PMID: 38587959 PMCID: PMC11374469 DOI: 10.1109/tmi.2024.3386108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Multimodal machine learning models are being developed to analyze pathology images and other modalities, such as gene expression, to gain clinical and biological insights. However, most frameworks for multimodal data fusion do not fully account for the interactions between different modalities. Here, we present an attention-based fusion architecture that integrates a graph representation of pathology images with gene expression data and concomitantly learns from the fused information to predict patient-specific survival. In our approach, pathology images are represented as undirected graphs, and their embeddings are combined with embeddings of gene expression signatures using an attention mechanism to stratify tumors by patient survival. We show that our framework improves the survival prediction of human non-small cell lung cancers, outperforming existing state-of-the-art approaches that leverage multimodal data. Our framework can facilitate spatial molecular profiling to identify tumor heterogeneity using pathology images and gene expression data, complementing results obtained from more expensive spatial transcriptomic and proteomic technologies.
Collapse
|
24
|
Travis WD, Eisele M, Nishimura KK, Aly RG, Bertoglio P, Chou TY, Detterbeck FC, Donnington J, Fang W, Joubert P, Kernstine K, Kim YT, Lievens Y, Liu H, Lyons G, Mino-Kenudson M, Nicholson AG, Papotti M, Rami-Porta R, Rusch V, Sakai S, Ugalde P, Van Schil P, Yang CFJ, Cilento VJ, Yotsukura M, Asamura H. The International Association for the Study of Lung Cancer (IASLC) Staging Project for Lung Cancer: Recommendation to Introduce Spread Through Air Spaces as a Histologic Descriptor in the Ninth Edition of the TNM Classification of Lung Cancer. Analysis of 4061 Pathologic Stage I NSCLC. J Thorac Oncol 2024; 19:1028-1051. [PMID: 38508515 DOI: 10.1016/j.jtho.2024.03.015] [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: 12/08/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Spread through air spaces (STAS) consists of lung cancer tumor cells that are identified beyond the edge of the main tumor in the surrounding alveolar parenchyma. It has been reported by meta-analyses to be an independent prognostic factor in the major histologic types of lung cancer, but its role in lung cancer staging is not established. METHODS To assess the clinical importance of STAS in lung cancer staging, we evaluated 4061 surgically resected pathologic stage I R0 NSCLC collected from around the world in the International Association for the Study of Lung Cancer database. We focused on whether STAS could be a useful additional histologic descriptor to supplement the existing ones of visceral pleural invasion (VPI) and lymphovascular invasion (LVI). RESULTS STAS was found in 930 of 4061 of the pathologic stage I NSCLC (22.9%). Patients with tumors exhibiting STAS had a significantly worse recurrence-free and overall survival in both univariate and multivariable analyses involving cohorts consisting of all NSCLC, specific histologic types (adenocarcinoma and other NSCLC), and extent of resection (lobar and sublobar). Interestingly, STAS was independent of VPI in all of these analyses. CONCLUSIONS These data support our recommendation to include STAS as a histologic descriptor for the Ninth Edition of the TNM Classification of Lung Cancer. Hopefully, gathering these data in the coming years will facilitate a thorough analysis to better understand the relative impact of STAS, LVI, and VPI on lung cancer staging for the Tenth Edition TNM Stage Classification.
Collapse
Affiliation(s)
- William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Megan Eisele
- Cancer Research And Biostatistics (CRAB), Seattle, Washington
| | | | - Rania G Aly
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pietro Bertoglio
- IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Teh-Ying Chou
- Department of Pathology and Laboratory Medicine, Taipei, Veterans General Hospital, Taipei, Taiwan
| | | | | | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Jiaotong University Medical School, Shanghai, People's Republic of China
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec - Université Laval, Quebec City, Canada
| | - Kemp Kernstine
- Department of Cardiovascular and Thoracic Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yolande Lievens
- Radiation Oncology, Ghent University Hospital and Ghent University, Gent, Belgium
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangdong, People's Republic of China
| | - Gustavo Lyons
- Buenos Aires British Hospital, Buenos Aires, Argentina
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton Hospital, London, United Kingdom
| | - Mauro Papotti
- Department of Oncology, University of Turin, Torino, Italy
| | - Ramon Rami-Porta
- Department of Thoracic Surgery, Hospital Universitari Mútua Terrassa, University of Barcelona, and CIBERES Lung Cancer Group, Terrassa, Barcelona, Spain
| | - Valerie Rusch
- Thoracic Surgery Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shuji Sakai
- Tokyo Women's Medical University, Tokyo, Japan
| | - Paula Ugalde
- Brigham & Women's Hospital, Boston, Massachusetts
| | - Paul Van Schil
- Antwerp University and Antwerp University Hospital, (Edegem) Antwerp, Belgium
| | - Chi-Fu Jeffrey Yang
- Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | | | - Masaya Yotsukura
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hisao Asamura
- Department of Thoracic Surgery, Keio University, Tokyo, Japan
| |
Collapse
|
25
|
Iwai M, Yokota E, Ishida Y, Yukawa T, Naomoto Y, Monobe Y, Haisa M, Takigawa N, Fukazawa T, Yamatsuji T. Establishment and characterization of novel high mucus-producing lung tumoroids derived from a patient with pulmonary solid adenocarcinoma. Hum Cell 2024; 37:1194-1204. [PMID: 38632190 PMCID: PMC11194211 DOI: 10.1007/s13577-024-01060-3] [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: 01/12/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024]
Abstract
Among mucus-producing lung cancers, invasive mucinous adenocarcinoma of the lung is a rare and unique subtype of pulmonary adenocarcinoma. Notably, mucus production may also be observed in the five subtypes of adenocarcinoma grouped under the higher-level diagnosis of Invasive Non-mucinous Adenocarcinomas (NMA). Overlapping pathologic features in mucus-producing tumors can cause diagnostic confusion with significant clinical consequences. In this study, we established lung tumoroids, PDT-LUAD#99, from a patient with NMA and mucus production. The tumoroids were derived from the malignant pleural effusion of a patient with lung cancer and have been successfully developed for long-term culture (> 11 months). Karyotyping by fluorescence in situ hybridization using an alpha-satellite probe showed that tumoroids harbored aneuploid karyotypes. Subcutaneous inoculation of PDT-LUAD#99 lung tumoroids into immunodeficient mice resulted in tumor formation, suggesting that the tumoroids were derived from cancer. Xenografts from PDT-LUAD#99 lung tumoroids reproduced the solid adenocarcinoma with mucin production that was observed in the patient's metastatic lymph nodes. Immunoblot analysis showed MUC5AC secretion into the culture supernatant of PDT-LUAD#99 lung tumoroids, which in contradistinction was barely detected in the culture supernatants of NCI-A549 and NCI-H2122 pulmonary adenocarcinoma cells known for their mucin-producing abilities. Here, we established a novel high-mucus-producing lung tumoroids from a solid adenocarcinoma. This preclinical model may be useful for elucidating the pathogenesis of mucus-producing lung cancer.
Collapse
Affiliation(s)
- Miki Iwai
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, Japan
| | - Etsuko Yokota
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | - Yuta Ishida
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | - Takuro Yukawa
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | - Yoshio Naomoto
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| | | | - Minoru Haisa
- Kawasaki Medical School General Medical Center, Okayama, Japan
- Department of Medical Care Work, Kawasaki College of Health Professions, Okayama, Japan
- Kawasaki Geriatric Medical Center, Okayama, Japan
| | - Nagio Takigawa
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, Japan
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, Japan
| | - Takuya Fukazawa
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, Japan.
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan.
| | - Tomoki Yamatsuji
- Department of General Surgery, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
26
|
Enfield KS, Colliver E, Lee C, Magness A, Moore DA, Sivakumar M, Grigoriadis K, Pich O, Karasaki T, Hobson PS, Levi D, Veeriah S, Puttick C, Nye EL, Green M, Dijkstra KK, Shimato M, Akarca AU, Marafioti T, Salgado R, Hackshaw A, TRACERx consortium, Jamal-Hanjani M, van Maldegem F, McGranahan N, Glass B, Pulaski H, Walk E, Reading JL, Quezada SA, Hiley CT, Downward J, Sahai E, Swanton C, Angelova M. Spatial Architecture of Myeloid and T Cells Orchestrates Immune Evasion and Clinical Outcome in Lung Cancer. Cancer Discov 2024; 14:1018-1047. [PMID: 38581685 PMCID: PMC11145179 DOI: 10.1158/2159-8290.cd-23-1380] [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: 11/17/2023] [Revised: 02/27/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024]
Abstract
Understanding the role of the tumor microenvironment (TME) in lung cancer is critical to improving patient outcomes. We identified four histology-independent archetype TMEs in treatment-naïve early-stage lung cancer using imaging mass cytometry in the TRACERx study (n = 81 patients/198 samples/2.3 million cells). In immune-hot adenocarcinomas, spatial niches of T cells and macrophages increased with clonal neoantigen burden, whereas such an increase was observed for niches of plasma and B cells in immune-excluded squamous cell carcinomas (LUSC). Immune-low TMEs were associated with fibroblast barriers to immune infiltration. The fourth archetype, characterized by sparse lymphocytes and high tumor-associated neutrophil (TAN) infiltration, had tumor cells spatially separated from vasculature and exhibited low spatial intratumor heterogeneity. TAN-high LUSC had frequent PIK3CA mutations. TAN-high tumors harbored recently expanded and metastasis-seeding subclones and had a shorter disease-free survival independent of stage. These findings delineate genomic, immune, and physical barriers to immune surveillance and implicate neutrophil-rich TMEs in metastasis. SIGNIFICANCE This study provides novel insights into the spatial organization of the lung cancer TME in the context of tumor immunogenicity, tumor heterogeneity, and cancer evolution. Pairing the tumor evolutionary history with the spatially resolved TME suggests mechanistic hypotheses for tumor progression and metastasis with implications for patient outcome and treatment. This article is featured in Selected Articles from This Issue, p. 897.
Collapse
Affiliation(s)
- Katey S.S. Enfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Claudia Lee
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Alastair Magness
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David A. Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Kristiana Grigoriadis
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Takahiro Karasaki
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
| | - Philip S. Hobson
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Dina Levi
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Clare Puttick
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Emma L. Nye
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Mary Green
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Krijn K. Dijkstra
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Masako Shimato
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ayse U. Akarca
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Teresa Marafioti
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Allan Hackshaw
- Cancer Research UK and University College London Cancer Trials Centre, London, United Kingdom
| | | | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Febe van Maldegem
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | | | | | | | - James L. Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Pre-cancer Immunology Laboratory, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Sergio A. Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Crispin T. Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Mihaela Angelova
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
27
|
Del Pino Herrera A, Ferrall-Fairbanks MC. A war on many fronts: cross disciplinary approaches for novel cancer treatment strategies. Front Genet 2024; 15:1383676. [PMID: 38873108 PMCID: PMC11169904 DOI: 10.3389/fgene.2024.1383676] [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: 04/26/2024] [Indexed: 06/15/2024] Open
Abstract
Cancer is a disease characterized by uncontrolled cellular growth where cancer cells take advantage of surrounding cellular populations to obtain resources and promote invasion. Carcinomas are the most common type of cancer accounting for almost 90% of cancer cases. One of the major subtypes of carcinomas are adenocarcinomas, which originate from glandular cells that line certain internal organs. Cancers such as breast, prostate, lung, pancreas, colon, esophageal, kidney are often adenocarcinomas. Current treatment strategies include surgery, chemotherapy, radiation, targeted therapy, and more recently immunotherapy. However, patients with adenocarcinomas often develop resistance or recur after the first line of treatment. Understanding how networks of tumor cells interact with each other and the tumor microenvironment is crucial to avoid recurrence, resistance, and high-dose therapy toxicities. In this review, we explore how mathematical modeling tools from different disciplines can aid in the development of effective and personalized cancer treatment strategies. Here, we describe how concepts from the disciplines of ecology and evolution, economics, and control engineering have been applied to mathematically model cancer dynamics and enhance treatment strategies.
Collapse
Affiliation(s)
- Adriana Del Pino Herrera
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Meghan C. Ferrall-Fairbanks
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
28
|
Chang JC, Rekhtman N. Pathologic Assessment and Staging of Multiple Non-Small Cell Lung Carcinomas: A Paradigm Shift with the Emerging Role of Molecular Methods. Mod Pathol 2024; 37:100453. [PMID: 38387831 PMCID: PMC11102290 DOI: 10.1016/j.modpat.2024.100453] [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: 12/05/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
Non-small cell lung carcinomas (NSCLCs) commonly present as 2 or more separate tumors. Biologically, this encompasses 2 distinct processes: separate primary lung carcinomas (SPLCs), representing independently arising tumors, and intrapulmonary metastases (IPMs), representing intrapulmonary spread of a single tumor. The advent of computed tomography imaging has substantially increased the detection of multifocal NSCLCs. The strategies and approaches for distinguishing between SPLCs and IPMs have evolved significantly over the years. Recently, genomic sequencing of somatic mutations has been widely adopted to identify targetable alterations in NSCLC. These molecular techniques have enabled pathologists to reliably discern clonal relationships among multiple NSCLCs in clinical practice. However, a standardized approach to evaluating and staging multiple NSCLCs using molecular methods is still lacking. Here, we reviewed the historical context and provided an update on the growing applications of genomic testing as a clinically relevant benchmark for determining clonal relationships in multiple NSCLCs, a practice we have designated "comparative molecular profiling." We examined the strengths and limitations of the morphology-based distinction of SPLCs vs IPMs and highlighted pivotal clinical and pathologic insights that have emerged from studying multiple NSCLCs using genomic approaches as a gold standard. Lastly, we suggest a practical approach for evaluating multiple NSCLCs in the clinical setting, considering the varying availability of molecular techniques.
Collapse
Affiliation(s)
- Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
29
|
Yang H, Liu X, Wang L, Zhou W, Tian Y, Dong Y, Zhou K, Chen L, Wang M, Wu H. 18 F-FDG PET/CT characteristics of IASLC grade 3 invasive adenocarcinoma and the value of 18 F-FDG PET/CT for preoperative prediction: a new prognostication model. Nucl Med Commun 2024; 45:338-346. [PMID: 38312089 DOI: 10.1097/mnm.0000000000001819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
OBJECTIVE This study is performed to investigate the imaging characteristics of the International Association for the Study of Lung Cancer grade 3 invasive adenocarcinoma (IAC) on PET/CT and the value of PET/CT for preoperative predicting this tumor. MATERIALS AND METHODS We retrospectively enrolled patients with IAC from August 2015 to September 2022. The clinical characteristics, serum tumor markers, and PET/CT features were analyzed. T test, Mann-Whitney U test, χ 2 test, Logistic regression analysis, and receiver operating characteristic analysis were used to predict grade 3 tumor and evaluate the prediction effectiveness. RESULTS Grade 3 tumors had a significantly higher maximum standardized uptake value (SUV max ) and consolidation-tumor-ratio (CTR) ( P < 0.001), while Grade 1 - 2 tumors were prone to present with air bronchogram sign or vacuole sign ( P < 0.001). A stepwise logistic regression analysis revealed that smoking history, CEA, SUV max , air bronchogram sign or vacuole sign and CTR were useful predictors for Grade 3 tumors. The established prediction model based on the above 5 parameters generated a high AUC (0.869) and negative predictive value (0.919), respectively. CONCLUSION Our study demonstrates that grade 3 IAC has a unique PET/CT imaging feature. The prognostication model established with smoking history, CEA, SUV max , air bronchogram sign or vacuole sign and CTR can effectively predict grade 3 tumors before the operation.
Collapse
Affiliation(s)
- Hanyun Yang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Manoutcharian K, Gevorkian G. Are we getting closer to a successful neoantigen cancer vaccine? Mol Aspects Med 2024; 96:101254. [PMID: 38354548 DOI: 10.1016/j.mam.2024.101254] [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: 10/24/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Although significant advances in immunotherapy have revolutionized the treatment of many cancer types over the past decade, the field of vaccine therapy, an important component of cancer immunotherapy, despite decades-long intense efforts, is still transmitting signals of promises and awaiting strong data on efficacy to proceed with regulatory approval. The field of cancer vaccines faces standard challenges, such as tumor-induced immunosuppression, immune response in inhibitory tumor microenvironment (TME), intratumor heterogeneity (ITH), permanently evolving cancer mutational landscape leading to neoantigens, and less known obstacles: neoantigen gain/loss upon immunotherapy, the timing and speed of appearance of neoantigens and responding T cell clonotypes and possible involvement of immune interference/heterologous immunity, in the complex interplay between evolving tumor epitopes and the immune system. In this review, we discuss some key issues related to challenges hampering the development of cancer vaccines, along with the current approaches focusing on neoantigens. We summarize currently well-known ideas/rationales, thus revealing the need for alternative vaccine approaches. Such a discussion should stimulate vaccine researchers to apply out-of-box, unconventional thinking in search of new avenues to deal with critical, often yet unaddressed challenges on the road to a new generation of therapeutics and vaccines.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| |
Collapse
|
31
|
Kolb T, Müller S, Möller P, Barth TFE, Marienfeld R. Molecular heterogeneity in histomorphologic subtypes of lung adeno carcinoma represents a challenge for treatment decision. Neoplasia 2024; 49:100955. [PMID: 38310709 PMCID: PMC10848034 DOI: 10.1016/j.neo.2023.100955] [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: 08/01/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024]
Abstract
Lung cancer is the leading cause in cancer related death, with non-small cell lung cancer (NSCLC) being the most frequent subtype. The importance of NSCLC is reflected by the various targeted therapy options especially for NSCLC adenocarcinomas (lung adeno carcinoma (LUAD)) as well as a set of options for immune therapies. However, despite these therapy advances, the majority of patients do not show a long-term response to either targeted therapy or immune checkpoint inhibition. One reason for treatment failure appears to be the NSCLC tumor heterogeneity. NSCLC heterogeneity might lead to an insufficient molecular characterization of a given sample due to the limited tumor material used for pathological assessment as the majority of analyses is performed on small biopsies. To get a more detailed insight into the tumor heterogeneity of NSCLC LUAD, especially in the light of its different histomorphological growth patterns, we analysed isolated NSCLC growth pattern areas and the corresponding entire tumor samples of a cohort of 31 NSLCS LUAD patients and compared their mutational landscape and their expression profiles. While significant differences of complex biomarkers, like tumor mutational burden (TMB) or microsatellite instability (MSI), were not detected between the five growth patterns -lepidic, papillary, micropapillary, acinar, and solid- we observed various subclonal mutations and copy number variants. Moreover, RNASeq analysis revealed growth pattern specific expression profiles affecting cellular processes like apoptosis, metastasis and proliferation. Taken together, our data provide novel insights into the tumor heterogeneity of LUAD required to overcome tumor heterogeneity related therapy resistance.
Collapse
Affiliation(s)
- Tobias Kolb
- Institute of Pathology, Ulm University, Ulm, Germany
| | - Sarah Müller
- Institute of Pathology, Ulm University, Ulm, Germany
| | - Peter Möller
- Institute of Pathology, Ulm University, Ulm, Germany
| | | | | |
Collapse
|
32
|
Pan X, AbdulJabbar K, Coelho-Lima J, Grapa AI, Zhang H, Cheung AHK, Baena J, Karasaki T, Wilson CR, Sereno M, Veeriah S, Aitken SJ, Hackshaw A, Nicholson AG, Jamal-Hanjani M, Swanton C, Yuan Y, Le Quesne J, Moore DA. The artificial intelligence-based model ANORAK improves histopathological grading of lung adenocarcinoma. NATURE CANCER 2024; 5:347-363. [PMID: 38200244 PMCID: PMC10899116 DOI: 10.1038/s43018-023-00694-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 11/21/2023] [Indexed: 01/12/2024]
Abstract
The introduction of the International Association for the Study of Lung Cancer grading system has furthered interest in histopathological grading for risk stratification in lung adenocarcinoma. Complex morphology and high intratumoral heterogeneity present challenges to pathologists, prompting the development of artificial intelligence (AI) methods. Here we developed ANORAK (pyrAmid pooliNg crOss stReam Attention networK), encoding multiresolution inputs with an attention mechanism, to delineate growth patterns from hematoxylin and eosin-stained slides. In 1,372 lung adenocarcinomas across four independent cohorts, AI-based grading was prognostic of disease-free survival, and further assisted pathologists by consistently improving prognostication in stage I tumors. Tumors with discrepant patterns between AI and pathologists had notably higher intratumoral heterogeneity. Furthermore, ANORAK facilitates the morphological and spatial assessment of the acinar pattern, capturing acinus variations with pattern transition. Collectively, our AI method enabled the precision quantification and morphology investigation of growth patterns, reflecting intratumoral histological transitions in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoxi Pan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khalid AbdulJabbar
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Jose Coelho-Lima
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anca-Ioana Grapa
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Hanyun Zhang
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Alvin Ho Kwan Cheung
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Juvenal Baena
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- AstraZeneca Computational Pathology, Munich, Germany
| | - Takahiro Karasaki
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Claire Rachel Wilson
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Hope Against Cancer and Leicester Experimental Cancer Medicine Centre, Leicester, UK
| | - Marco Sereno
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Selvaraju Veeriah
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sarah J Aitken
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Andrew G Nicholson
- Department of Histopathology, Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Yinyin Yuan
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - John Le Quesne
- Molecular Pathology, School of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Cancer Research UK Beatson Institute of Cancer Research, Glasgow, UK.
- NHS Greater Glasgow and Clyde, Glasgow, UK.
| | - David A Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Department of Cellular Pathology, University College London Hospitals, London, UK.
| |
Collapse
|
33
|
Rolfo C, Russo A. Navigating into a stormy sea: liquid biopsy enters peri-operative management in early-stage non-small cell lung cancer. Ann Oncol 2024; 35:147-149. [PMID: 38331558 DOI: 10.1016/j.annonc.2023.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024] Open
Affiliation(s)
- C Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - A Russo
- Department of Onco-Hematology, Papardo Hospital, Messina, Italy
| |
Collapse
|
34
|
Xie L, Kong H, Yu J, Sun M, Lu S, Zhang Y, Hu J, Du F, Lian Q, Xin H, Zhou J, Wang X, Powell CA, Hirsch FR, Bai C, Song Y, Yin J, Yang D. Spatial transcriptomics reveals heterogeneity of histological subtypes between lepidic and acinar lung adenocarcinoma. Clin Transl Med 2024; 14:e1573. [PMID: 38318637 PMCID: PMC10844893 DOI: 10.1002/ctm2.1573] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Patients who possess various histological subtypes of early-stage lung adenocarcinoma (LUAD) have considerably diverse prognoses. The simultaneous existence of several histological subtypes reduces the clinical accuracy of the diagnosis and prognosis of early-stage LUAD due to intratumour intricacy. METHODS We included 11 postoperative LUAD patients pathologically confirmed to be stage IA. Single-cell RNA sequencing (scRNA-seq) was carried out on matched tumour and normal tissue. Three formalin-fixed and paraffin-embedded cases were randomly selected for 10× Genomics Visium analysis, one of which was analysed by digital spatial profiler (DSP). RESULTS Using DSP and 10× Genomics Visium analysis, signature gene profiles for lepidic and acinar histological subtypes were acquired. The percentage of histological subtypes predicted for the patients from samples of 11 LUAD fresh tissues by scRNA-seq showed a degree of concordance with the clinicopathologic findings assessed by visual examination. DSP proteomics and 10× Genomics Visium transcriptomics analyses revealed that a negative correlation (Spearman correlation analysis: r = -.886; p = .033) between the expression levels of CD8 and the expression trend of programmed cell death 1(PD-L1) on tumour endothelial cells. The percentage of CD8+ T cells in the acinar region was lower than in the lepidic region. CONCLUSIONS These findings illustrate that assessing patient histological subtypes at the single-cell level is feasible. Additionally, tumour endothelial cells that express PD-L1 in stage IA LUAD suppress immune-responsive CD8+ T cells.
Collapse
Affiliation(s)
- Linshan Xie
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Hui Kong
- Department of PathologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Jinjie Yu
- Department of Thoracic SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Department of Thoracic SurgeryShanghai Geriatric Medical CenterShanghaiChina
| | - Mengting Sun
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Shaohua Lu
- Department of PathologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Yong Zhang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Jie Hu
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
| | - Fang Du
- Department of AnesthesiologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Qiuyu Lian
- Gurdon InstituteUniversity of CambridgeCambridgeUK
| | - Hongyi Xin
- Global Institute of Future TechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Jian Zhou
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Engineer and Technology Research Center of Internet of Things for Respiratory MedicineShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Respiratory Research InstitutionShanghaiChina
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
- Shanghai Engineering Research for AI Technology for Cardiopulmonary DiseasesFudan University Shanghai Medical CollegeShanghaiChina
| | - Charles A. Powell
- Pulmonary, Critical Care and Sleep MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Fred R. Hirsch
- Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health SystemNew YorkNew YorkUSA
| | - Chunxue Bai
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Engineer and Technology Research Center of Internet of Things for Respiratory MedicineShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Respiratory Research InstitutionShanghaiChina
| | - Yuanlin Song
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Engineer and Technology Research Center of Internet of Things for Respiratory MedicineShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Respiratory Research InstitutionShanghaiChina
| | - Jun Yin
- Department of Thoracic SurgeryZhongshan HospitalFudan UniversityShanghaiChina
| | - Dawei Yang
- Department of Pulmonary and Critical Care MedicineZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Engineer and Technology Research Center of Internet of Things for Respiratory MedicineShanghaiChina
- Shanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Shanghai Respiratory Research InstitutionShanghaiChina
- Department of Pulmonary and Critical Care MedicineZhongshan Hospital (Xiamen)Fudan UniversityXiamenChina
| |
Collapse
|
35
|
Rodriguez-Quintero JH, Kamel MK, Jindani R, Vimolratana M, Chudgar NP, Stiles BM. Sublobar resection is associated with less lymph nodes examined and lower delivery of adjuvant therapy in patients with 1.5- to 2.0-cm clinical IA2 non-small-cell lung cancer: a retrospective cohort study. Eur J Cardiothorac Surg 2024; 65:ezad431. [PMID: 38147358 PMCID: PMC11007732 DOI: 10.1093/ejcts/ezad431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/11/2023] [Accepted: 12/25/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVES CALGB140503, in which nodal sampling was mandated, reported non-inferior disease-free survival for patients undergoing sublobar resection (SLR) compared to lobectomy (L). Outside of trial settings, the adequacy of lymphadenectomy during SLR has been questioned. We sought to evaluate whether SLR is associated with suboptimal lymphadenectomy, differences in pathologic upstaging and survival in patients with 1.5- to 2.0-cm tumours using real-world data. MATERIALS AND METHODS Using the National Cancer Database(2018-2019), we evaluated patients with 1.5- to 2.0-cm non-small-cell lung cancer who underwent resection (sublobar versus lobectomy). We studied factors associated with nodal upstaging (logistic regression) and survival (Cox regression and Kaplan-Meier method) after propensity matching to adjust for differences among groups. RESULTS Among 3196 patients included, SLR was performed in 839 (26.3%) (of which 588 were wedge resections) and L was performed in 2357 (73.7%) patients. More patients undergoing SLR (21.7%) compared to L (2.1%) had no lymph nodes sampled (P < 0.001). Those undergoing SLR had fewer total lymph nodes examined (4 vs 11, P < 0.001) and were less likely to have pathologic nodal metastases (4.7% vs 9%, P < 0.001) compared to L. Multivariable analysis identified L [adjusted odds ratio (aOR) 2.21, 95% confidence interval, 1.47-3.35] to be independently associated with pathologic N+ disease. Overall survival was not associated with the type of procedure but was significantly decreased in those with N+ disease. CONCLUSIONS Despite comparable overall survival to L, SLR is associated with suboptimal lymphadenectomy in patients with 1.5-2.0 cm non-small-cell lung cancer. Surgeons should be careful to perform adequate lymphadenectomy when performing SLR to mitigate nodal under-staging and to identify appropriate patients for systemic therapy.
Collapse
Affiliation(s)
| | - Mohamed K Kamel
- Department of Cardiothoracic Surgery, University of Rochester Medical
Center, Rochester, NY, USA
| | - Rajika Jindani
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical
Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marc Vimolratana
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical
Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Neel P Chudgar
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical
Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Brendon M Stiles
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical
Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
36
|
LIU B. [Hypothesis of Genetic Diversity Selection in the Occurrence and Development of
Lung Cancer: Molecular Evolution and Clinical Significance]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 26:943-949. [PMID: 38163980 PMCID: PMC10767663 DOI: 10.3779/j.issn.1009-3419.2023.101.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Indexed: 01/03/2024]
Abstract
So far, the monoclonal hypothesis of tumor occurrence and development cannot be justified. The genetic diversity selection hypothesis for the occurrence and development of lung cancer links Mendelian genetics with Darwin's theory of evolution, suggesting that the genetic diversity of tumor cell populations with polyclonal origins-monoclonal selection-subclonal expansion is the result of selection pressure. Normal cells acquire mutations in oncogenic driver genes and have a selective advantage over other cells, becoming tumor initiating cells; In the interaction with the tumor microenvironment (TME), the vast majority of initiating cells are recognized and killed by the human immune system. If immune escape occurs, the incidence of malignant tumors will greatly increase, and subclonal expansion, intratumour heterogeneity, etc. will occur. This article proposed the hypothesis of genetic diversity selection and analyzed its clinical significance.
.
Collapse
|
37
|
Pyo JS, Lee BH, Min KW, Kim NY. Clinicopathological significances of cribriform pattern in lung adenocarcinoma. Pathol Res Pract 2024; 253:155035. [PMID: 38171080 DOI: 10.1016/j.prp.2023.155035] [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: 11/15/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
The present study aimed to investigate the clinicopathological and prognostic implications of the cribriform pattern in lung adenocarcinoma through a meta-analysis. The estimated rates of cribriform pattern in lung adenocarcinomas were investigated. The correlations between cribriform pattern and clinicopathological characteristics, including genetic alterations and prognosis were evaluated. The estimated rate of cribriform pattern was 0.150 (95% confidence interval [CI], 0.101-0.218) in lung adenocarcinoma. The estimated rates of cribriform pattern in the 5% and 10% criteria were 0.230 (95% CI 0.125-0.386) and 0.130 (95% CI 0.062-0.252), respectively. The presence of cribriform pattern was significantly correlated with larger tumor size (> 30 mm), spread through air spaces, and lymph node metastasis (P < 0.001, P < 0.001, and P = 0.007, respectively, in the meta-regression test). There were no significant differences between cribriform pattern, smoking history, and vascular and lymphatic invasion. In lung adenocarcinoma with cribriform pattern, the estimated rates of ALK rearrangement, KRAS, and EGFR mutations were 0.407 (95% CI 0.165-0.704), 0.330 (95% CI 0.117-0.646), and 0.249 (95% CI 0.125-0.437), respectively. ALK rearrangement was significantly more frequent in lung adenocarcinomas with cribriform pattern than in those without. The overall survival rate was significantly worse in lung adenocarcinomas with a cribriform pattern than in those without (hazard ratio 2.051, 95% CI 1.369-3.075). In conclusion, the presence of a cribriform pattern can be a useful predictor of the clinicopathological characteristics and prognosis of patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Byoung-Hoon Lee
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Nae Yu Kim
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
38
|
Grigoriadis K, Huebner A, Bunkum A, Colliver E, Frankell AM, Hill MS, Thol K, Birkbak NJ, Swanton C, Zaccaria S, McGranahan N. CONIPHER: a computational framework for scalable phylogenetic reconstruction with error correction. Nat Protoc 2024; 19:159-183. [PMID: 38017136 DOI: 10.1038/s41596-023-00913-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 08/24/2023] [Indexed: 11/30/2023]
Abstract
Intratumor heterogeneity provides the fuel for the evolution and selection of subclonal tumor cell populations. However, accurate inference of tumor subclonal architecture and reconstruction of tumor evolutionary histories from bulk DNA sequencing data remains challenging. Frequently, sequencing and alignment artifacts are not fully filtered out from cancer somatic mutations, and errors in the identification of copy number alterations or complex evolutionary events (e.g., mutation losses) affect the estimated cellular prevalence of mutations. Together, such errors propagate into the analysis of mutation clustering and phylogenetic reconstruction. In this Protocol, we present a new computational framework, CONIPHER (COrrecting Noise In PHylogenetic Evaluation and Reconstruction), that accurately infers subclonal structure and phylogenetic relationships from multisample tumor sequencing, accounting for both copy number alterations and mutation errors. CONIPHER has been used to reconstruct subclonal architecture and tumor phylogeny from multisample tumors with high-depth whole-exome sequencing from the TRACERx421 dataset, as well as matched primary-metastatic cases. CONIPHER outperforms similar methods on simulated datasets, and in particular scales to a large number of tumor samples and clones, while completing in under 1.5 h on average. CONIPHER enables automated phylogenetic analysis that can be effectively applied to large sequencing datasets generated with different technologies. CONIPHER can be run with a basic knowledge of bioinformatics and R and bash scripting languages.
Collapse
Affiliation(s)
- Kristiana Grigoriadis
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Abigail Bunkum
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Lab, University College London Cancer Institute, London, UK
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| | - Simone Zaccaria
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK.
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
39
|
Li Z, Yin Z, Luan Z, Zhang C, Wang Y, Zhang K, Chen F, Yang Z, Tian Y. Comprehensive analyses for the coagulation and macrophage-related genes to reveal their joint roles in the prognosis and immunotherapy of lung adenocarcinoma patients. Front Immunol 2023; 14:1273422. [PMID: 38022584 PMCID: PMC10644034 DOI: 10.3389/fimmu.2023.1273422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose This study aims to explore novel biomarkers related to the coagulation process and tumor-associated macrophage (TAM) infiltration in lung adenocarcinoma (LUAD). Methods The macrophage M2-related genes were obtained by Weighted Gene Co-expression Network Analysis (WGCNA) in bulk RNA-seq data, while the TAM marker genes were identified by analyzing the scRNA-seq data, and the coagulation-associated genes were obtained from MSigDB and KEGG databases. Survival analysis was performed for the intersectional genes. A risk score model was subsequently constructed based on the survival-related genes for prognosis prediction and validated in external datasets. Results In total, 33 coagulation and macrophage-related (COMAR) genes were obtained, 19 of which were selected for the risk score model construction. Finally, 10 survival-associated genes (APOE, ARRB2, C1QB, F13A1, FCGR2A, FYN, ITGB2, MMP9, OLR1, and VSIG4) were involved in the COMAR risk score model. According to the risk score, patients were equally divided into low- and high-risk groups, and the prognosis of patients in the high-risk group was significantly worse than that in the low-risk group. The ROC curve indicated that the risk score model had high sensitivity and specificity, which was validated in multiple external datasets. Moreover, the model also had high efficacy in predicting the clinical outcomes of LUAD patients who received anti-PD-1/PD-L1 immunotherapy. Conclusion The COMAR risk score model constructed in this study has excellent predictive value for the prognosis and immunotherapeutic clinical outcomes of patients with LUAD, which provides potential biomarkers for the treatment and prognostic prediction.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, China
| | - Zongxiu Yin
- Department of Pulmonary and Critical Care Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zupeng Luan
- Department of Radiation Oncology, Jinan Third People’s Hospital, Jinan, China
| | - Chi Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanyuan Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kai Zhang
- Generalsurgery Department, Wen-shang County People’s Hospital, Wenshang, China
| | - Feng Chen
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhensong Yang
- Department of Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yuan Tian
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, China
| |
Collapse
|
40
|
Garbo E, Del Rio B, Ferrari G, Cani M, Napoli VM, Bertaglia V, Capelletto E, Rolfo C, Novello S, Passiglia F. Exploring the Potential of Non-Coding RNAs as Liquid Biopsy Biomarkers for Lung Cancer Screening: A Literature Review. Cancers (Basel) 2023; 15:4774. [PMID: 37835468 PMCID: PMC10571819 DOI: 10.3390/cancers15194774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Lung cancer represent the leading cause of cancer mortality, so several efforts have been focused on the development of a screening program. To address the issue of high overdiagnosis and false positive rates associated to LDCT-based screening, there is a need for new diagnostic biomarkers, with liquid biopsy ncRNAs detection emerging as a promising approach. In this scenario, this work provides an updated summary of the literature evidence about the role of non-coding RNAs in lung cancer screening. A literature search on PubMed was performed including studies which investigated liquid biopsy non-coding RNAs biomarker lung cancer patients and a control cohort. Micro RNAs were the most widely studied biomarkers in this setting but some preliminary evidence was found also for other non-coding RNAs, suggesting that a multi-biomarker based liquid biopsy approach could enhance their efficacy in the screening context. However, further studies are needed in order to optimize detection techniques as well as diagnostic accuracy before introducing novel biomarkers in the early diagnosis setting.
Collapse
Affiliation(s)
- Edoardo Garbo
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Benedetta Del Rio
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Giorgia Ferrari
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Massimiliano Cani
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Valerio Maria Napoli
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Valentina Bertaglia
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Enrica Capelletto
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Health System, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, 10124 Orbassano, Italy; (E.G.); (B.D.R.); (G.F.); (M.C.); (V.M.N.); (V.B.); (E.C.); (S.N.)
| |
Collapse
|
41
|
Lorenz C, Hillmer AM, Brägelmann J. Predicting the next move: tracking the complexity of lung cancer evolution and metastasis. Signal Transduct Target Ther 2023; 8:291. [PMID: 37553337 PMCID: PMC10409755 DOI: 10.1038/s41392-023-01567-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Affiliation(s)
- Carina Lorenz
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Mildred Scheel School of Oncology Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
| | - Axel M Hillmer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Pathology, Cologne, Germany
| | - Johannes Brägelmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Mildred Scheel School of Oncology Cologne, Cologne, Germany.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany.
| |
Collapse
|
42
|
Szeitz B, Glasz T, Herold Z, Tóth G, Balbisi M, Fillinger J, Horváth S, Mohácsi R, Kwon HJ, Moldvay J, Turiák L, Szász AM. Spatially Resolved Proteomic and Transcriptomic Profiling of Anaplastic Lymphoma Kinase-Rearranged Pulmonary Adenocarcinomas Reveals Key Players in Inter- and Intratumoral Heterogeneity. Int J Mol Sci 2023; 24:11369. [PMID: 37511126 PMCID: PMC10380216 DOI: 10.3390/ijms241411369] [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: 06/09/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Pulmonary adenocarcinomas (pADCs) with an ALK rearrangement are a rare cancer subtype, necessitating comprehensive molecular investigations to unravel their heterogeneity and improve therapeutic strategies. In this pilot study, we employed spatial transcriptomic (NanoString GeoMx) and proteomic profiling to investigate seven treatment-naïve pADCs with an ALK rearrangement. On each FFPE tumor slide, 12 smaller and 2-6 larger histopathologically annotated regions were selected for transcriptomic and proteomic analysis, respectively. The correlation between proteomics and transcriptomics was modest (average Pearson's r = 0.43 at the gene level). Intertumoral heterogeneity was more pronounced than intratumoral heterogeneity, and normal adjacent tissue exhibited distinct molecular characteristics. We identified potential markers and dysregulated pathways associated with tumors, with a varying extent of immune infiltration, as well as with mucin and stroma content. Notably, some markers appeared to be specific to the ALK-driven subset of pADCs. Our data showed that within tumors, elements of the extracellular matrix, including FN1, exhibited substantial variability. Additionally, we mapped the co-localization patterns of tumor microenvironment elements. This study represents the first spatially resolved profiling of ALK-driven pADCs at both the gene and protein expression levels. Our findings may contribute to a better understanding of this cancer type prior to treatment with ALK inhibitors.
Collapse
Affiliation(s)
- Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.)
| | - Tibor Glasz
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary
| | - Zoltán Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.)
| | - Gábor Tóth
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Mirjam Balbisi
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Pharmaceutical Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - János Fillinger
- Department of Pathology, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Szabolcs Horváth
- Department of Pathology, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Réka Mohácsi
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.)
| | - Ho Jeong Kwon
- Department of Biotechnology, Division of Life Sciences, Yonsei University, Seoul 03722, Republic of Korea
| | - Judit Moldvay
- 1st Department of Pulmonology, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Lilla Turiák
- MS Proteomics Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Pharmaceutical Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Marcell Szász
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.)
- Department of Tumor Biology, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
43
|
Martínez-Ruiz C, Black JRM, Puttick C, Hill MS, Demeulemeester J, Larose Cadieux E, Thol K, Jones TP, Veeriah S, Naceur-Lombardelli C, Toncheva A, Prymas P, Rowan A, Ward S, Cubitt L, Athanasopoulou F, Pich O, Karasaki T, Moore DA, Salgado R, Colliver E, Castignani C, Dietzen M, Huebner A, Al Bakir M, Tanić M, Watkins TBK, Lim EL, Al-Rashed AM, Lang D, Clements J, Cook DE, Rosenthal R, Wilson GA, Frankell AM, de Carné Trécesson S, East P, Kanu N, Litchfield K, Birkbak NJ, Hackshaw A, Beck S, Van Loo P, Jamal-Hanjani M, Swanton C, McGranahan N. Genomic-transcriptomic evolution in lung cancer and metastasis. Nature 2023; 616:543-552. [PMID: 37046093 PMCID: PMC10115639 DOI: 10.1038/s41586-023-05706-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/04/2023] [Indexed: 04/14/2023]
Abstract
Intratumour heterogeneity (ITH) fuels lung cancer evolution, which leads to immune evasion and resistance to therapy1. Here, using paired whole-exome and RNA sequencing data, we investigate intratumour transcriptomic diversity in 354 non-small cell lung cancer tumours from 347 out of the first 421 patients prospectively recruited into the TRACERx study2,3. Analyses of 947 tumour regions, representing both primary and metastatic disease, alongside 96 tumour-adjacent normal tissue samples implicate the transcriptome as a major source of phenotypic variation. Gene expression levels and ITH relate to patterns of positive and negative selection during tumour evolution. We observe frequent copy number-independent allele-specific expression that is linked to epigenomic dysfunction. Allele-specific expression can also result in genomic-transcriptomic parallel evolution, which converges on cancer gene disruption. We extract signatures of RNA single-base substitutions and link their aetiology to the activity of the RNA-editing enzymes ADAR and APOBEC3A, thereby revealing otherwise undetected ongoing APOBEC activity in tumours. Characterizing the transcriptomes of primary-metastatic tumour pairs, we combine multiple machine-learning approaches that leverage genomic and transcriptomic variables to link metastasis-seeding potential to the evolutionary context of mutations and increased proliferation within primary tumour regions. These results highlight the interplay between the genome and transcriptome in influencing ITH, lung cancer evolution and metastasis.
Collapse
Affiliation(s)
- Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare Puttick
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Jonas Demeulemeester
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
- Integrative Cancer Genomics Laboratory, Department of Oncology, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Elizabeth Larose Cadieux
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
- Medical Genomics, University College London Cancer Institute, London, UK
| | - Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Thomas P Jones
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Antonia Toncheva
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Paulina Prymas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Laura Cubitt
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Foteini Athanasopoulou
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Carla Castignani
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
- Medical Genomics, University College London Cancer Institute, London, UK
| | - Michelle Dietzen
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Miljana Tanić
- Medical Genomics, University College London Cancer Institute, London, UK
- Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Emilia L Lim
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Ali M Al-Rashed
- Centre for Nephrology, Division of Medicine, University College London, London, UK
| | - Danny Lang
- Scientific Computing STP, Francis Crick Institute, London, UK
| | - James Clements
- Scientific Computing STP, Francis Crick Institute, London, UK
| | - Daniel E Cook
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Rachel Rosenthal
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Gareth A Wilson
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
| | | | - Philip East
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Stephan Beck
- Medical Genomics, University College London Cancer Institute, London, UK
| | - Peter Van Loo
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals, London, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute and University College London Cancer Institute, London, UK.
- Department of Medical Oncology, University College London Hospitals, London, UK.
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|